Previous Article in Journal
Molecular Interactions at Lipid Droplet–Mitochondria Membrane Contact Sites in Mammalian Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dysregulated Lipid Metabolism as a Central Driver of Atherosclerotic Plaque Pathology

by
Julia Emily Steinbeck
1,†,
Rachel Anne Iannotti
1,† and
Adil Rasheed
1,2,*
1
Immunology Center of Georgia, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
2
Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Lipidology 2025, 2(4), 17; https://doi.org/10.3390/lipidology2040017
Submission received: 16 August 2025 / Revised: 26 September 2025 / Accepted: 27 September 2025 / Published: 30 September 2025

Abstract

It has long been recognized that elevated circulating lipid levels are among the strongest risk factors for the development of plaques within the arterial wall that are characteristic of atherosclerotic cardiovascular disease. Indeed, decades of studies have identified the deposition of low-density lipoprotein as an initiator of this disease, which coordinates the vascular and immune dysfunction that fuels the advancement of the atherosclerotic plaque. However, in the vessel wall, deposited cholesterol and fatty acids are dynamic in nature and engage signaling pathways. Shifting from metabolic-related pathways, lipid modifications and their conversion to intermediates engage signaling cascades that further perpetuate the inflammatory milieu of the atherosclerotic plaque and its progression towards the fatal end-stage events associated with cardiovascular disease, including myocardial infarction. In this review, we will cover the cellular and molecular mechanisms that preserve homeostasis and advance disease, including how lipid species induce endothelial dysfunction and drive the development of macrophage foam cells. We will additionally discuss ongoing therapeutic strategies to combat the hyperlipidemia that underlies atherogenesis.

1. Introduction

Lipids, including triglycerides, phospholipids, and cholesterol, are a diverse set of macromolecules that play important roles in preserving cellular homeostasis [1,2,3]. Their production, transport, and metabolism are all involved in critical biological processes, such as production of adenosine triphosphate (ATP) for energy, components of cell membranes, synthesis of hormones, and cell signaling, among many other functions. However, when these processes go awry, diseases such as atherosclerotic cardiovascular disease (ASCVD) develop. Otherwise referred to as ischemic heart disease or coronary artery disease (CAD), ASCVD is a complication of cardiovascular disease (CVD) and remains the number one cause of mortality worldwide [4,5,6]. This disease can occur sub-clinically for several decades as lipids deposit within the arterial wall to form the plaques characteristic of ASCVD. As the plaque advances through sequential stages, it becomes vulnerable and prone to rupture or erosion, leading to major adverse cardiovascular events (MACE), including stroke or myocardial infarction [7,8,9,10]. Traditional risk factors for ASCVD include age, genetics, diabetes and other metabolic diseases, hypertension, smoking, and elevated low-density lipoprotein (LDL) cholesterol [11]. Apart from simple accumulation in the artery, evolving studies have described the critical influences of lipoprotein and lipid subtypes in signaling and the advancement of the plaque towards these end-stage complications. In this review, we will cover the production and transport of lipids and their contribution within the arterial wall towards driving atherosclerotic plaque pathology. We will also highlight existing and new lipid-based therapeutic targets for the treatment of ASCVD.

2. Hepatic Synthesis, Composition, and Transport of Circulating Lipoproteins

Lipoproteins are lipid transport molecules that support the movement of hydrophobic lipids through the circulation to peripheral tissues [1]. Lipoproteins include chylomicrons, very low-density lipoprotein (VLDL), intermediate-density lipoprotein (IDL), low-density lipoprotein (LDL), and high-density lipoprotein (HDL). While IDL, LDL, and HDL are considered cholesterol-rich lipoproteins, chylomicrons and VLDL are triglyceride-rich lipoproteins (TRLs). These lipoproteins are produced by the liver, except for chylomicrons, which are assembled in the small intestine. In addition to their major lipid species and origin, lipoproteins are classified based on several parameters, including size and density, which have been reviewed extensively elsewhere [2,12]. VLDL composition includes various apolipoproteins embedded in the phospholipid monolayer, such as apolipoprotein-B100 and -C3 (Apo-B100 and Apo-C3, respectively), which serve as mediators of lipoprotein biology [13,14]. Tissue metabolism of VLDL by the adipose tissue and skeletal muscle is mediated by lipoprotein, hepatic, and endothelial lipases, which liberate triglycerides. Removal of these triglycerides from VLDL particles can then form IDL and LDL [1,13,14]. Additionally, hydrolysis of TRLs by lipoprotein lipase (LPL) decreases particle size, allowing them to more easily cross the endothelium, where they can be retained as part of the atherosclerotic plaque [15,16,17].
LDL cholesterol was first recognized as a major driver of atherosclerosis in 1955 [18,19,20]. Synthesis of LDL by the liver includes one Apo-B100 per LDL particle, which is a glycoprotein synthesized and secreted by the liver [2]. Colloquially referred to as the “bad cholesterol”, LDL is responsible for the transport of cholesterol from the liver to peripheral tissues, including the arterial wall. LDL in circulation is cleared by the liver via binding of Apo-B100 on the LDL particle to low-density lipoprotein receptor (LDLR) in the liver. Upon binding, LDL-LDLR is internalized by hepatocytes via receptor-mediated endocytosis, where LDL is trafficked to the lysosome for degradation [20,21]. Subsequently, LDLR is returned to the plasma membrane to promote further clearance of LDL from the circulation. Despite being cloned and sequenced in the mid-1980s, the structure of Apo-B100 on LDL and interactions with LDLR were only described in 2024 [22,23,24,25]. These interactions of Apo-B100 with LDLR are critical for the uptake and clearance of LDL cholesterol from the circulation and therefore may open further therapeutic avenues for patients who require more aggressive lipid-lowering.
HDL, on the other hand, is often referred to as the “good cholesterol”. The major apolipoprotein on HDL is Apo-A1. Unlike LDL, which carries only one Apo-B100, there are multiple Apo-A1 per HDL particle [2,26]. HDL has been found to have many roles that participate in atherogenesis, which are reviewed elsewhere [27]. The role of HDL as the “good cholesterol” in atherosclerosis relates to reverse cholesterol transport (RCT), which begins in the plaque where Apo-A1 and HDL receive cholesterol via cholesterol efflux, followed by transport to the liver [28]. Apo-A1 on HDL subsequently binds to scavenger receptor B1 (SR-B1) expressed on hepatocytes, which then engulf the HDL particle. Considered the rate-limiting enzyme of RCT, cholesterol 7 alpha-hydroxylase (CYP7A1) converts cholesterol to bile acids, and its efflux is mediated by obligate heterodimers of adenosine triphosphate-binding cassette transporter G5/G8 (ABCG5/8). This ultimately leads to the excretion of cholesterol in the feces via the intestine. In fact, a recent systems biology approach using metabolomics and transcriptomics in pig models of atherosclerosis has identified elevations in bile acids in driving inter-organ dysfunction leading to atherogenesis [29].
Given the importance of these lipoproteins in the development of atherosclerosis, levels of these lipid species have been established as a diagnostic measure in assessing ASCVD risk (Table 1). Of clinical importance, familial hypercholesterolemia (FH) is characterized by chronic elevations in LDL cholesterol, which affects approximately 1 in 220 people worldwide [30,31]. This condition is underscored by genetic variants in LDLR, Apo-B, or gain-of-function mutations in proprotein convertase subtilisin/kexin type 9 (PCSK9), all of which function to prevent the clearance of LDL cholesterol from the circulation.

3. Lipid-Dependent Aortic Endothelial Dysfunction and Monocyte Entry

Lipid-mediated dysfunction, as one of the central drivers of ASCVD, is captured by three prevailing hypotheses. The first hypothesis put forth by Ross and Glomset is the response-to-injury hypothesis, which states that endothelial injury due to hyperlipidemia, among other insults, initiates lipid accumulation in the tissue and subsequently the inflammation that drives atherosclerosis [37,38]. Secondly, the cumulative exposure hypothesis postulates that modified LDL becomes trapped within the intima of the artery, causing initial plaque development, which in turn increases shear stress and mechanical burden of the vessel wall [39]. Through continuous LDL accumulation, there is further dysfunction of the plaque that drives progression towards MACE. The third hypothesis is the lipid retention hypothesis, whereby excess lipoproteins cross the endothelium, enter the arterial wall, and are then subject to modifications, such as oxidation, leading to oxidized low-density lipoprotein (oxLDL) or aggregation, yielding aggregated low-density lipoprotein (agLDL) [40]. These modified LDLs subsequently induce the formation of macrophage foam cells, discussed below, which in turn lead to the cooperativity of lipids and inflammation that potentiate plaque development [41]. Below, we discuss the direct effects that occur in response to lipids, where the effects of disturbed flow secondary to plaque growth and feedback mechanisms that further induce atherogenic signaling have been reviewed extensively elsewhere [42,43].
Owing to its role in vascular surveillance and providing a physical barrier between the circulation and the tissue, it is unsurprising that a unifying mechanism in the development of ASCVD is changes to the endothelium, including its activation, dysfunction, and mesenchymal transition [44,45]. Briefly, endothelial activation involves the increased expression of pro-inflammatory adhesion molecules, cytokines, and chemokines. Endothelial dysfunction is characterized by impaired nitric oxide signaling and redox imbalance. Atherogenic lipids can also cause endothelial cells to adopt a mesenchymal phenotype, termed endothelial-to-mesenchymal transition (EndMT) [46]. However, these endothelial defects do not occur as separate processes. For example, nitric oxide itself has been shown to repress inflammatory signaling in part by blocking NF-κB signaling through S-nitrosylation of NF-κB subunits or inhibitory κB kinase [47,48,49,50]. Nevertheless, this results in blunted downstream activation of endothelial cells, including production of inflammatory cytokines and adhesion molecules, as described below.
One of the hallmarks of a dysfunctional endothelium is increased permeability due to structural changes that compromise barrier integrity [51]. In fact, hypercholesterolemia and atherogenic lipids have been shown to inhibit the production of nitric oxide from endothelial cells via promoting inhibitory interactions in the plasma membrane between caveolin and the enzyme that produces nitric oxide, endothelial nitric oxide synthase, thereby impairing endothelial function [52,53,54,55]. Passive diffusion has long been touted as the mechanism by which circulating LDL can enter the artery due to increased endothelial permeability [56]. However, more recent studies have challenged this notion and proposed an alternative mechanism for LDL transport. Known as transcytosis, LDL is transported from the lumen to the intima across the endothelial cells. There are two forms of transcytosis, fluid-phase and receptor-mediated transcytosis [57,58]. In the former, caveolae facilitate the uptake of LDL at the cell surface, whereas in the latter, LDL uptake is mediated by cell surface receptors, including LDLR and SR-B1. Of note, expression of SR-B1 and its partner DOCK4 (dedicator of cytokinesis 4) have been found to be increased in mouse and human atherosclerosis-prone aortic regions [59]. Interestingly, as an additional anti-atherogenic function, Apo-A1 on HDL competes with LDL for SR-B1 binding and thus reduces the transcytosis of LDL across the endothelium and its subsequent accumulation in the aorta [60].
Endothelial cell activation involves pro-inflammatory processes that promote leukocyte extravasation into the plaque (Figure 1). Common to these pro-inflammatory mechanisms that drive atherogenesis, oxLDL, which is a prominent modified LDL particle found in human atheromas, activates the inflammatory transcription factor NF-κB in macrophages as well as endothelial cells [61,62,63]. OxLDL also increases expression of one of its receptors, lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1, also known as OLR1), to promote NF-κB nuclear translocation and subsequent pro-inflammatory signaling [64,65,66]. One of the key lipid-induced pro-inflammatory processes of endothelial cells that drives atherogenesis is the induction of selectins, such as P- and E-selectin, that slow leukocyte rolling along the endothelium [67,68,69,70,71,72,73]. Furthermore, endothelial cells also upregulate adhesion molecules, including vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1), although studies in mice suggest a greater importance of VCAM-1, particularly in early atherosclerosis [74,75,76,77,78,79]. These adhesion molecules, their respective ligands, and signaling pathways have been reviewed extensively elsewhere [67,73,80]. Importantly, upregulation of these factors on endothelial cells induced by oxLDL and LOX-1 are reversed by cholesterol-lowering treatment (statins) [81].
Of importance to plaque development, observations in mice and humans have robustly demonstrated increased circulating monocytes during the hyperlipidemia that underscores ASCVD [82,83,84,85]. Broadly, monocytes are divided into three subsets: classical (Ly6Chi in mice, CD14+ CD16 in humans), intermediate (Ly6Cint in mice, CD14+ CD16+ in humans), and nonclassical monocytes (Ly6Clo in mice, CD14dim CD16+ in humans) [86,87,88,89]. In particular, hyperlipidemia favors the production of classical monocytes, which are pro-inflammatory in nature. Observations in mice demonstrate that hypercholesterolemia supports the survival and proliferation of these classical monocytes, which is reversed upon treatment with statins [90]. Furthermore, the spleen is an important reservoir for the production of classical monocytes during atherosclerosis [91]. This is correlated with increased splenic activity as determined by functional imaging in patients with CVD, which was used as a prognostic indicator for future MACE [92]. In addition to their production, hypercholesterolemia also promotes the chemotaxis of monocytes towards the injured vessel wall in part due to an upregulation of chemokine receptors, such as C-C motif chemokine receptor 2 (CCR2) [93,94,95]. Mechanistic studies demonstrate that in response to oxLDL, endothelial cells produce chemokines to recruit these monocytes, including monocyte chemotactic protein 1 (MCP-1, also known as chemokine (C-C motif) ligand 2 [CCL2]) and interleukin-8 (IL-8, also known as CXCL8) [96,97,98,99,100]. Specifically, deletion of these factors or their cognate receptors demonstrates their roles in driving atherogenesis [101,102,103].

4. Macrophage Scavenging and Metabolism of Lipids

Once in the lesion, monocytes encounter macrophage colony-stimulating factor (M-CSF, also referred to as colony-stimulating factor 1 [CSF1]), recognized by their receptor CSF1 receptor, which stimulates their differentiation into macrophages to promote plaque development [104,105,106]. In fact, within the plaque, monocytes are short-lived and readily differentiated into macrophages within 24 h [90]. As part of the lipid-induced dysfunction that drives atherogenesis, oxLDL also induces endothelial expression of M-CSF [99,107]. Although M-CSF can be produced by numerous cell types, conditional knockout studies have identified smooth muscle and endothelial cells, but not monocytes/macrophages, as producers of M-CSF responsible for monocyte differentiation [108]. In turn, these macrophages scavenge cholesterol-rich lipoproteins that have deposited in the arterial wall to become the foam cells characteristic of atherosclerosis [104,109]. Owing to the modification of LDL that occurs within the plaque microenvironment, macrophages depend on expression of scavenger receptors for the uptake of oxLDL. The increased negativity of LDL resulting from its oxidation increases its binding affinity to scavenger receptors, and its retention is supported by binding to proteoglycans, which together increase oxLDL uptake by macrophages and ultimately foam cell genesis within the plaque [110,111,112,113].
The most prevalent of these scavenger receptors responsible for modified lipoprotein uptake is the cluster of differentiation 36 (CD36) and scavenger receptor class A (SR-A). CD36 signals via assembly into a heterotrimeric signaling complex with toll-like receptors 2/4/6 (TLR2/4/6), whereas SR-A signals through the homo-trimeric complex [114,115,116,117,118]. The engagement of these receptors by oxLDL initiates extensive signaling mechanisms that promote the production of pro-inflammatory cytokines, oxLDL uptake, and cell death, all of which increase the pro-inflammatory microenvironment of the plaque [119,120,121]. In addition, macrophages also express LOX-1, which, under basal conditions, accounts for a relatively low amount of oxLDL uptake (~5–10%) but is markedly increased upon stimulation and upregulation of LOX-1 [122]. CD36 was first discovered in human monocytes when precipitated with the OKM5 antibody [123]. Since then, the role of CD36 in atherosclerosis has been controversial. In some studies, CD36 knockout mice showed a decrease in foam cell formation coupled with decreased total cholesterol levels [124,125]. However, other studies have shown the opposite, with CD36 or SR-A knockout mice having increased cholesterol levels with increased or no change in lesion area [126]. Furthermore, double knockout of CD36 and SR-A in mice improves features of atherosclerosis progression, such as death and necrotic area, without impacting overall lesion area or lipid parameters [127]. In addition to differences in numerous variables, these studies serve to underscore the complexity of these receptors apart from their roles in scavenging lipids and the need to better understand the lipid-inflammatory axis in ASCVD. Apart from oxLDL-mediated signaling, TLR4 participates in the catabolism of agLDL for uptake in macrophages in cooperation with CD14 in a MyD88-dependent pathway involving lysosomal exocytosis [128]. Apart from receptor-mediated lipoprotein uptake, a recent study has identified macropinocytosis, a form of fluid-phase uptake of macromolecules mediated by membrane ruffling, as contributing to foam cell development in both murine and human atheromas [129].
Regardless of the route of engulfment, ingested lipids undergo endocytic trafficking to the lysosome for metabolism, where free cholesterol is liberated from the lipoprotein via hydrolysis by lysosomal acid lipase (LAL) [130]. However, free cholesterol is cytotoxic to the cell and must be metabolized or eliminated to preserve cellular homeostasis. In the endoplasmic reticulum (ER), free cholesterol is esterified to fatty acids by Acyl-CoA:cholesterol acyltransferase 1 (ACAT1) to produce cholesteryl esters, which are safely stored in lipid droplets. These lipid droplets can be catabolized by a process known as autophagy to liberate free cholesterol and fatty acids [131]. This free cholesterol can then be removed from the macrophages by cholesterol efflux, which is the first step in RCT [28]. ABCA1 and ABCG1, known as cholesterol efflux transporters, remove free cholesterol to the extracellular compartment via their acceptors, Apo-A1 and HDL, respectively [28,132,133]. In the cell, cholesterol can also be converted to oxysterols by enzymes such as cytochrome P450 enzymes or hydroxylases, or by non-enzymatic means such as reactive oxygen species [134]. Oxysterols such as 22-, 24-, 25-, or 27-hydroxycholesterol serve as endogenous ligands for the liver x receptors α/β (LXRα/β), which have been shown to have anti-atherogenic effects [135,136,137,138,139,140]. Belonging to the nuclear receptor superfamily, LXRs are activated by these oxysterols to act as transcription factors to promote cholesterol homeostasis, including the induction of genes involved in RCT, such as ABCA1, ABCG1, and CYP7A1. Of note, mutations in ABCA1 lead to Tangier disease, a genetic condition resulting in the accumulation of cholesteryl esters in various tissues and low levels of serum HDL [141,142]. Furthermore, LXRs also repress inflammatory gene transcription through antagonism of transcription factors like NF-κB or cis-repression of inflammatory genes [143,144], which together reduce atherogenesis. Additionally, intermediates of cholesterol synthesis, such as desmosterol, also induce signaling in macrophages to dampen inflammation and repress atherosclerosis development [145,146]. In addition to macrophages, a significant proportion of foam cells are derived from smooth muscle cells in both murine and human atheromas [147,148,149,150]. Indeed, several of the aforementioned mechanisms of lipid handling are defective in these foam cell subsets. This includes low levels of LAL for hydrolysis of cholesteryl esters and their limited capacity to engage autophagy of lipid droplets, subsequently impairing cholesterol efflux [151,152].
However, in the setting of excess lipid content within the atherosclerotic plaque, these homeostatic mechanisms go awry, leading to macrophage foam cell dysfunction and their active contribution to the advancement of this disease. One of the hallmarks of foam cells is endoplasmic reticulum stress, including the unfolded protein response (UPR). This consists of PKR-like eukaryotic initiation factor 2A kinase (PERK), activating transcription factor 6 (ATF6), inositol-requiring enzyme 1 (IRE1), as well as activation of CCAAT-enhancer-binding protein homologous protein (CHOP), which together function to promote lipid-induced cell death [104,153,154]. Apart from intracellular cholesterol stores, the formation of cholesterol-rich inflammarafts at the plasma membrane allows for the assembly of inflammatory receptor dimers and engagement of downstream inflammatory signaling [155,156]. Although under steady-state conditions, 25-hydroxycholesterol promotes LXR activation to mitigate intracellular cholesterol stores, in the setting of excess cholesterol accumulation, recent studies show 25-hydroxycholesterol accumulation in human coronary atheromas [157]. Importantly, independent of LXR activation, 25-hydroxycholesterol produced by macrophages increases vascular inflammation through reorganization of membrane cholesterol and TLR4 signaling, in addition to promoting adverse plaque remodeling.
Fatty acids also play critical roles in driving the pathogenesis of atherosclerosis. As previously mentioned, autophagy, which is involved in the hydrolysis of cholesteryl esters, also releases free fatty acids [131]. Fatty acids in macrophages can also be produced by de novo fatty acid synthesis, autophagy of lipid droplets, or can be taken up from the extracellular compartment via scavenger receptors [158,159,160]. In fact, the metabolism of fatty acids has been shown to drive macrophage phenotypes, influencing atherosclerotic plaque pathology. Many studies have shown that the balance between fatty acid oxidation (catabolic) and synthesis (anabolic) induces an anti-inflammatory (M2-like) or pro-inflammatory (M1-like) phenotype of macrophages, respectively, thus influencing overall atherosclerotic plaque pathology [160,161,162,163].

5. Fatty Acids as Signaling Molecules

Apart from serving as metabolic substrates, fatty acids and their derivatives also act as signaling molecules. Fatty acid diversity encompasses carbon chain length and degree of saturation, which can be saturated, monounsaturated, or polyunsaturated. Saturated fatty acids are prevalent and drive chronic inflammatory diseases, including atherosclerosis, via engagement of CD36 and TLRs to promote downstream inflammasome activation [118,160,164]. A prominent example of such a long-chain saturated fatty acid with profound effects on atherosclerosis is palmitate. In addition to metabolism, fatty acids such as palmitate can also serve as substrates for post-translational modifications. In particular, palmitoylation of CD36 and LOX-1 promotes receptor processing and localization to lipid rafts, half-life, and oxLDL uptake to drive foam cell accumulation [165,166].
Additionally, polyunsaturated fatty acids (PUFAs) have been extensively studied in the context of atherosclerosis, in particular omega-3 and omega-6 (ω-3 and ω-6, respectively) PUFAs [167]. A prevalent example of these signaling fatty acid species is arachidonic acid (AA), which is a polyunsaturated omega-6 fatty acid that provides fluidity to cell membranes [168]. Relevant to atherosclerosis, engulfment of oxLDL and subsequent metabolism produces AA in macrophage foam cells [169,170]. AA can be produced via metabolism from linoleic acid, which is an essential ω-6 PUFA that cannot be synthesized by mammals, or by AA release from cell membranes by phospholipases. Regardless of the route of production, AA can be metabolized to produce eicosanoids via enzymatic means, including i) prostaglandins and thromboxanes produced by cyclooxygenases (COX), ii) leukotrienes produced by lipoxygenases, and iii) hydroeicosatetraenoic acids (HETEs) or epoxyeicosatetraenoic acids (EETs) by cytochrome P450 enzymes [168]. These metabolites and enzymes all play varying roles in ASCVD [170].
However, of greater interest to ASCVD are ω-3 PUFAs, which represent another class of bioactive fatty acid species and are synthesized from α-linolenic acid, an essential fatty acid found in vegetable and fish oils, seeds, and nuts [171]. In general, several epidemiologic studies have associated increased consumption of ω-3 PUFAs, such as fish, with a lower risk of CVD [172,173,174]. Among the most relevant ω-3 PUFAs to CVD are eicosapentaenoic acid (EPA), which is synthesized from α-linolenic acid, and docosahexaenoic acid (DHA), which is produced by conversion from EPA. EPA, in particular, has been the subject of several clinical trials wherein the Japan EPA Lipid Intervention Study (JELIS) and the Reduction of Cardiovascular Events with Isocapent Ethyl Intervention Trial (REDUCE-IT) studies have both shown that EPA lowers the risk of CVD events [175,176].
EPA and DHA can be further metabolized into a class of molecules known as specialized pro-resolving mediators (SPMs), including lipoxins, maresins, protectins, and resolvins [177,178]. Here, we will focus on resolvins, which are the SPMs that have been best studied in atherosclerosis. The E-series of resolvins is derived from EPA, of which resolving E1 (RvE1) is the most relevant in atherosclerosis. RvE1 binds to ChemR23, a G protein-coupled receptor (GPCR) whose engagement promotes efferocytosis and dampens macrophage inflammation [179,180]. In contrast, DHA produces maresins, protectins, as well as the D-series of resolvins [177,178]. Resolvin D1 (RvD1), in particular, has been the most well-studied of this series in atherosclerosis. RvD1 binds to a GPR32, a GPCR, and an ALX/FPR2 receptor. Overall, RvD1 and its receptor have also been shown to have atheroprotective roles by promoting efferocytosis and dampening inflammation [178,181,182,183].

6. Other Lipid Species as Drivers of Atherosclerosis

Although cholesterol-rich lipoproteins were initially recognized as conferring risk for ASCVD, recent studies have shown that TRLs are also causal agents of atherosclerosis, including studies demonstrating that TRLs on a per particle basis are more atherogenic than LDL [16,184,185,186]. In addition to Apo-B, TRLs also possess Apo-C3, which in both type 1 and type 2 diabetic patients positively correlates with the risk of developing ASCVD, with loss-of-function mutations associated with a reduction in circulating triglycerides and atherosclerosis [187,188,189,190,191,192,193]. Mechanistic studies have attributed these deleterious roles of Apo-C3 to increased formation of VLDL by the liver and inhibiting lipoprotein lipase-mediated lipolysis of VLDL, thereby blunting its metabolism and maintaining high TRL circulating levels [194,195,196]. Though best studied in the context of TRL, Apo-C3 is also present on LDL and contributes to its atherogenic effects [197].
As indicated in Table 1, lipoprotein (a) (Lp(a)) is another critical lipoprotein species that confers ASCVD risk [198,199]. Lp(a) is a modified form of LDL, where the apolipoprotein(a) glycoprotein is covalently linked to Apo-B100 via a disulfide bond [200,201]. Interestingly, recent genetic evidence using Mendelian randomization demonstrated that Apo-B associated with Lp(a) has a greater hazard ratio for atherosclerosis than LDL-ApoB [202,203]. Lp(a) also contains oxidized phospholipids (discussed below), which account for some of its atherogenic effects [204,205]. In addition, Lp(a) supports leukocyte extravasation into the plaque by interacting with monocyte integrins and stimulating endothelial cell release of chemokines and expression of adhesion molecules to support monocyte-endothelial interactions [206,207]. Indeed, macrophage foam cells can also uptake Lp(a), which promotes apoptosis through CD36-TLR2 signaling [118,208].
Another lipid species that has received decades of attention for its role in atherogenesis is ceramides, which consist of a fatty acid conjugated to sphingosine. The biology of ceramides and their in-depth metabolic pathways have been extensively reviewed elsewhere [209,210], where cellular levels of ceramides are regulated by breakdown from sphingomyelin found in the cell membrane, de novo biosynthesis, or ceramide catabolism. Given the drastic roles this class of sphingolipids plays in disease, ceramides have been used as a clinical biomarker for a variety of diseases, including diabetes and CVD [211]. Owing to the variability in fatty acid species, as described above, in humans, specific ceramide subspecies found in circulation have been associated with increased CVD risk [212,213,214,215,216,217]. Ceramides are found enriched in LDL and therefore can be deposited in the arterial wall. Furthermore, increased circulating levels of these ceramides are associated with increased plaque pathology, including vulnerability and necrotic core content, leading to future MACE [218,219]. In mice, strategies to increase ceramides by targeting metabolism, as described above, have demonstrated an overall pro-atherogenic effect through promoting lipoprotein transport and retention in the plaque, LDL aggregation, and monocyte adhesion [220,221,222,223,224].
In addition to cholesterol, oxidized phospholipids (oxPLs) found on lipoproteins also act as signaling lipids to induce atherogenesis [225]. Of importance, oxPL-ApoB levels show a greater correlation with Lp(a) rather than oxLDL in circulation, likely due to the stronger association of oxPLs in Lp(a) [204,226]. Furthermore, oxPL-ApoB levels show greater predictive value for future CVD as compared to Lp(a) levels, further impressing the importance of oxPL in atherogenesis [205]. oxPLs are a heterogeneous species, where over 30 different oxPLs have been identified in atherosclerotic plaques [227]. Beyond association studies, the pathogenicity of oxPLs was demonstrated in an atherogenic mouse expressing a natural antibody to (E06-scFV), which neutralizes oxPL signaling, resulting in a reduction in circulating lipid levels and overall atherosclerotic plaque area [228]. Neutralizing oxPLs dampens macrophage inflammatory status, apoptosis, and scavenger receptor signaling [118,228,229,230,231]. Two prominent examples of oxPLs investigated in the pathogenesis of atherosclerosis are 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC) and oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (oxPAPC), which accumulate in the plaque to induce cell death in the vessel wall, regulatory T cell dysfunction, and pro-inflammatory responses in monocytes and macrophages [232,233,234,235,236,237,238].

7. Lipids as Drivers of Non-Apoptotic Cell Death

One of the key features of the advanced or vulnerable plaque is the development of a necrotic core, which is an acellular region that develops as cells shift towards pro-inflammatory modes of cell death and ultimately leads to the fatal complications associated with ASCVD [239]. Although best described in the context of inflammation, here we will highlight the lipid-dependent mechanisms by which these processes occur and drive plaque pathology. While necrosis occurs in a signaling-independent manner, necroptosis also results in cell membrane lysis but importantly relies on a cell signaling cascade. This notably involves the phosphorylation of necrosome components, receptor-interacting serine/threonine-protein kinases 1 and 3 (RIPK1 and 3), leading to the activation of mixed lineage kinase domain-like protein (MLKL) and its insertion into the membrane [239]. Necroptosis has been identified as a process that occurs in both mouse and human atheromas and can be engaged by oxLDL [240]. Furthermore, silencing RIPK1, RIPK3, or MLKL or blocking necroptotic signaling have all demonstrated the importance of necroptotic cell death to the development of the necrotic core during atherosclerosis [240,241,242,243]. Importantly, necroptotic cells release lipid mediators, such as prostanoids, that further promote the inflammatory milieu of the plaque in part by blunting the clearance of dead cells [182].
Pyroptotic cell death also involves the formation of pores in the plasma membrane [239]. This form of cell death is characterized by the assembly and activation of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome and production of IL-1β, which is released through pores formed by proteins known as gasdermins, particularly gasdermin D [244]. The inflammasome involved in pyroptosis has been identified in human atheromas in the vicinity of the necrotic core and is been strongly associated with ASCVD [245,246,247]. Cholesterol crystals, which accumulate as a result of excess free cholesterol content, are found in the necrotic core and drive lysosomal damage and the activation of the NLRP3 inflammasome [115,248]. Importantly, gasdermin D must be cleaved by caspase-1 to allow for the N-terminal portion to participate in pore formation. However, recent evidence has shown that conjugation of the fatty acid palmitate (S-palmitoylation) to either intact or cleaved gasdermin D at Cys191 also promotes its membrane translocation, although this was shown to be more efficient in cleaved gasdermin D [249,250]. This lipid-dependent activation of the inflammasome, as described here for pyroptosis, also stimulates NETosis, whereby neutrophils release extracellular traps rich in DNA and enzymes [239,251].
Ferroptosis is characterized by lipid peroxide species via the peroxidation of PUFAs that occurs in an iron-dependent manner, which in turn disrupts the plasma membrane, leading to pore formation and cell death [239]. Furthermore, the metabolism of these lipid peroxides can form further lipid species that contribute to cellular toxicity. Interestingly, this form of cell death also contributes to LDL oxidation and thus the pathogenicity of this lipoprotein [252,253]. Indications of ferroptosis in human atheromas include the presence of iron-rich foam cells, hemoglobin from erythrophagocytosis, and ferroptotic regulators ferritin and heme oxygenase 1 (HMOX1) [254,255]. In mice, administration of the ferroptosis inhibitor ferrostatin-1 decreased overall lesion area and cell death, which was demonstrated to be driven in part by oxLDL-mediated lipid peroxidation and endothelial cell dysfunction [256].

8. Lipid Therapies in Atherosclerotic Cardiovascular Disease

Undoubtedly, with the overwhelming evidence of lipids as central drivers of ASCVD, it is unsurprising that targeting dyslipidemia remains an intensively investigated avenue for clinical intervention (Figure 2). Given the primary role of the liver in synthesizing and exporting lipoproteins, many lipid-based strategies for intervening in ASCVD involve targeting the liver. The best example are statins, which remain the most widely prescribed class of drugs in the world [257]. First discovered in 1976 and approved for use in 1987, statins inhibit HMG-CoA reductase, which is the rate-limiting enzyme in cholesterol biosynthesis in the liver, by competitive inhibition [20]. Across numerous studies, statins have been found to lower circulating cholesterol levels, leading to lower all-cause mortality and MACE, as well as being associated with reductions in plaque area and improved plaque stability [258,259,260]. Despite the success of statins, approximately 10–15% of patients develop statin intolerance [261,262]. Therefore, additional avenues of lipid lowering are currently being investigated to improve outcomes. Similar to statins, bempedoic acid has arisen as a recently approved drug to lower cholesterol biosynthesis in the liver [263]. Functioning as an inhibitor of the ATP citrate lyase (ACLY) enzyme, bempedoic acid blocks the conversion of citrate to acetyl-CoA, which serves as the first substrate in the cholesterol biosynthetic pathway. This therefore arrests cholesterol synthesis upstream of HMG-CoA reductase. In patients receiving the maximum tolerated statin dose or those who were statin intolerant, bempedoic acid led to a 16.5–21.1% reduction in LDL cholesterol compared to placebo, which was accompanied by a 13% reduction in MACE [264,265]. Another strategy to lower circulating cholesterol levels has been to target ApoB, which is a critical component of LDL and therefore also reduces LDL cholesterol levels. Mipomersen, an antisense oligonucleotide targeting Apo-B100, was initially approved for human use in patients with FH due to reductions in circulating LDL cholesterol and Lp(a); however, this approval was withdrawn due to hepatotoxicity concerns [266,267,268,269]. A more recent advance for targeting hepatic lipid metabolism is PCSK9, which prevents LDLR recycling to the membrane to support LDL clearance from the circulation, thereby inducing its degradation and promoting sustained hypercholesterolemia [270]. Currently, two monoclonal antibodies (Alirocumab and Evolocumab) and one small interfering RNA (Inclisiran) are approved for clinical use, which show reductions in circulating cholesterol levels and improved hazard ratios for primary cardiovascular endpoints [271,272,273,274,275]. Furthermore, Inclisiran was also shown to improve LDL cholesterol levels in patients with heterozygous FH [276]. In a recent multi-center clinical trial, it was found that Evolocumab reduced the lipid-rich necrotic core in the carotid bifurcation of patients with established but asymptomatic carotid stenosis [277]. Analysis of the ODYSSEY Outcomes clinical trial, which tested Alirocumab in ASCVD, revealed that baseline Lp(a) levels predicted future MACE and that there were beneficial reductions in Lp(a) levels with PCSK9 inhibition [278]. This has sparked multiple phase three clinical trials to test the effect of small interfering RNA and antisense oligonucleotide approaches to disrupting Lp(a) production by the liver on CVD outcomes [279].
Since the initial discovery of LDL cholesterol as a major risk factor of ASCVD, subsequent clinical cohorts have identified elevations in circulating triglyceride levels associated with CVD risk [15]. Based on the mechanisms described above, Apo-C3 has risen as a clinical target to reduce circulating triglyceride levels. Two approved antisense oligonucleotides targeting Apo-C3, Volanesorsen and Olezarsen, have shown reductions in circulating triglycerides, Apo-B, and non-HDL cholesterol [280,281,282]. Other classes of proteins being investigated for their triglyceride-reducing properties are angiopoietin-like proteins 3, 4, and 8 (ANGPTL3, 4, 8), which are LPL inhibitors produced by the liver [283]. Together, these ANGPTLs inhibit metabolism and increase circulating triglyceride levels. Although antibody and gene silencing approaches are being investigated to target these ANGPTLs, only the ANGPTL3 monoclonal antibody Evinacumab has been approved for patients with persistent hypercholesterolemia, such as those with homozygous FH, which decreases circulating LDL cholesterol levels [284,285].
Apart from targeting hepatic production of lipoproteins, another therapeutic strategy is to directly target the lipoproteins that mediate these effects in circulation. Owing to associations between low HDL cholesterol levels and CVD events, as well as the well-known role for HDL in RCT as well as other anti-atherogenic processes, there has been significant interest in efforts to boost HDL as a therapeutic measure to combat ASCVD [286]. However, Mendelian randomization studies to explore the causal effects of HDL on ASCVD, along with a variety of drugs to raise HDL cholesterol, have challenged this “HDL hypothesis” with many of these studies showing null effects on improving cardiovascular outcomes (reviewed in [286]). A prominent example of a class of HDL cholesterol-boosting drugs is cholesteryl ester transfer protein (CETP) inhibitors. CETP is responsible for the transfer of triglycerides from VLDL and LDL to HDL in exchange for cholesteryl esters, effectively raising LDL cholesterol and decreasing HDL cholesterol, thus providing a lipoprotein profile associated with atherogenesis [287,288]. Of the four phase three clinical trials that have investigated CETP inhibition to raise HDL cholesterol, three have failed to show improvements in CVD outcomes. However, one recent trial has shown positive results with respect to risk reduction in the first coronary event during the 4-year follow-up [289]. Newer approaches to address raising HDL in circulation include the administration of recombinant HDL (rHDL) and Apo-A1, which in animal models have shown removal of cholesterol from the plaque, as well as plaque regression (reviewed in [290]). However, the most recent AEGIS-II phase three clinical trial, which investigated the infusion of purified Apo-A1 in patients after acute myocardial infarction [291], did not show improvement in MACE up to 1-year post-treatment.

9. Conclusions

Despite emerging studies that identify additional risk for ASCVD, and even after more than 70 years of intense investigation, it remains undeniable that lipids, as a diverse species, play critical roles in the pathogenesis of the atherosclerotic plaque. Beginning with observations and early therapeutic strategies to target the production and accumulation of lipids, primarily cholesterol, in the arterial wall, a wealth of subsequent studies has identified these lipids as active biomolecules in driving disease progression. Alternatively, significant interest has grown in harnessing the beneficial aspects of lipid biology to promote disease regression. Although statins have proven effective at reducing cholesterol levels, there remains a lipid-dependent influence on ASCVD. Nevertheless, new treatments on the horizon illustrate the benefit of further investigation into lipid-lowering therapies and potential effects on downstream signaling pathways to improve overall cardiovascular health.

Author Contributions

Conceptualization, J.E.S., R.A.I. and A.R.; resources, A.R.; data curation, J.E.S., R.A.I. and A.R.; writing—original draft preparation, J.E.S., R.A.I. and A.R.; writing—review and editing, J.E.S., R.A.I. and A.R.; visualization, J.E.S., R.A.I. and A.R.; supervision, A.R.; project administration, A.R.; funding acquisition, A.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

All figures were created using BioRender.com.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AAArachidonic acid
ABCA1/G1/G5/G8Adenosine triphosphate-binding cassette transporter A1/G1/G5/G8
ACAT1Acetyl-CoA acetyltransferase 1
ACLYATP citrate lyase
agLDLAggregated LDL
ANGPTL3/4/8Angiopoietin-like 3/4/8
Apo-A1/B100/C3/(a)Apolipoprotein A1/B100/C3/(a)
ASCVDAtherosclerotic cardiovascular disease
ATF6Activating transcription factor 6
ATPAdenosine triphosphate
CADCoronary artery disease
CCL2/MCP-1Chemokine (C-C motif) ligand 2/Monocyte chemoattractant protein 1
CCR2C-C chemokine receptor type 2
CD36Cluster of differentiation 36
CETPCholesteryl ester transfer protein
CHOPCCAAT-enhancer-binding protein homologous protein
COXCyclooxygenase
CVDCardiovascular disease
CYP7A1Cholesterol 7 alpha-hydroxylase
DHADocosahexaenoic acid
DOCK4Dedicator of cytokinesis 4
EETEpoxyeicosatetraenoic acid
EndMTEndothelial-to-mesenchymal transition
EPAEicosapentaenoic acid
EREndoplasmic reticulum
FHFamilial hypercholesterolemia
GPCRG protein-coupled receptor
HETEHydroeicosatetraenoic acid
HDLHigh-density lipoprotein
HMOX1Heme oxygenase 1
ICAM-1Intercellular cell adhesion molecule 1
IDLIntermediate-density lipoprotein
IL-1β/8Interleukin-1β/8
IRE1Inositol-requiring enzyme 1
LALLysosomal acid lipase
LDLLow-density lipoprotein
LDLRLow-density lipoprotein receptor
LOX-1/OLR1Lectin-like oxidized low-density lipoprotein receptor 1/OxLDL receptor 1
Lp(a)Lipoprotein(a)
LPLLipoprotein lipase
LXRα/βLiver x receptor α/β
M-CSF/CSF1Macrophage-colony stimulating factor/Colony-stimulating factor 1
MACEMajor adverse cardiovascular events
MLKLMixed lineage kinase domain-like protein
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NLRP3NOD-, LRR- and pyrin domain-containing protein 3
oxLDLOxidized low-density lipoprotein
oxPAPCOxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine
oxPLOxidized phospholipids
PCSK9Proprotein convertase subtilisin/kexin type 9
PERKPKR-like eukaryotic initiation factor 2A kinase
POVPC1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine
PUFAPolyunsaturated fatty acid
RCTReverse cholesterol transport
rHDLRecombinant HDL
RIPK1/3Receptor-interacting serine/threonine-protein kinase 1/3
RvD1/E1Resolvin D1/E1
SPMSpecialized pro-resolving mediators
SR-A/B1Scavenger receptor class A/class B member 1
TLR2/4/6Toll-like receptor 2/4/6
TRLTriglyceride-rich lipoprotein
UPRUnfolded protein response
VCAM-1Vascular cell adhesion molecule 1
VLDLVery low-density lipoprotein

References

  1. Ahmed, S.; Shah, P.; Ahmed, O. Biochemistry, Lipids. In StatPearls; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
  2. Feingold, K.R. Introduction to Lipids and Lipoproteins. In Endotext; Feingold, K.R., Ahmed, S.F., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., de Herder, W.W., Dhatariya, K., Dungan, K., et al., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
  3. Yoon, H.; Shaw, J.L.; Haigis, M.C.; Greka, A. Lipid metabolism in sickness and in health: Emerging regulators of lipotoxicity. Mol. Cell 2021, 81, 3708–3730. [Google Scholar] [CrossRef]
  4. World Health Organization. Global Health Estimates: Life Expectancy and Leading Causes of Death and Disability. Available online: https://www.who.int/data/gho/data/themes/mortality-and-global-health-estimates (accessed on 7 August 2025).
  5. Nowbar, A.N.; Gitto, M.; Howard, J.P.; Francis, D.P.; Al-Lamee, R. Mortality From Ischemic Heart Disease. Circ. Cardiovasc. Qual. Outcomes 2019, 12, e005375. [Google Scholar] [CrossRef]
  6. Martin, S.S.; Aday, A.W.; Allen, N.B.; Almarzooq, Z.I.; Anderson, C.A.M.; Arora, P.; Avery, C.L.; Baker-Smith, C.M.; Bansal, N.; Beaton, A.Z.; et al. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025, 151, e41–e660. [Google Scholar] [CrossRef] [PubMed]
  7. Stary, H.C.; Chandler, A.B.; Dinsmore, R.E.; Fuster, V.; Glagov, S.; Insull, W., Jr.; Rosenfeld, M.E.; Schwartz, C.J.; Wagner, W.D.; Wissler, R.W. A definition of advanced types of atherosclerotic lesions and a histological classification of atherosclerosis. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation 1995, 92, 1355–1374. [Google Scholar] [CrossRef]
  8. Stary, H.C. Natural history and histological classification of atherosclerotic lesions: An update. Arter. Thromb. Vasc. Biol. 2000, 20, 1177–1178. [Google Scholar] [CrossRef] [PubMed]
  9. Libby, P.; Pasterkamp, G.; Crea, F.; Jang, I.K. Reassessing the Mechanisms of Acute Coronary Syndromes. Circ. Res. 2019, 124, 150–160. [Google Scholar] [CrossRef] [PubMed]
  10. Vergallo, R.; Park, S.J.; Stone, G.W.; Erlinge, D.; Porto, I.; Waksman, R.; Mintz, G.S.; D’Ascenzo, F.; Seitun, S.; Saba, L.; et al. Vulnerable or High-Risk Plaque: A JACC: Cardiovascular Imaging Position Statement. JACC Cardiovasc. Imaging 2025, 18, 709–740. [Google Scholar] [CrossRef]
  11. Arnett, D.K.; Blumenthal, R.S.; Albert, M.A.; Buroker, A.B.; Goldberger, Z.D.; Hahn, E.J.; Himmelfarb, C.D.; Khera, A.; Lloyd-Jones, D.; McEvoy, J.W.; et al. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019, 140, e563–e595. [Google Scholar] [CrossRef]
  12. Sorci-Thomas, M.G.; Hegele, R.A.; Remaley, A.T. A Century of Milestones and Breakthroughs Related to Low- and High-Density Lipoproteins. Arter. Thromb. Vasc. Biol. 2024, 44, 7–11. [Google Scholar] [CrossRef]
  13. Juarez Casso, F.M.; Farzam, K. Biochemistry, Very Low Density Lipoprotein. In StatPearls; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
  14. Chen, J.; Fang, Z.; Luo, Q.; Wang, X.; Warda, M.; Das, A.; Oldoni, F.; Luo, F. Unlocking the mysteries of VLDL: Exploring its production, intracellular trafficking, and metabolism as therapeutic targets. Lipids Health Dis. 2024, 23, 14. [Google Scholar] [CrossRef]
  15. Chapman, M.J.; Ginsberg, H.N.; Amarenco, P.; Andreotti, F.; Boren, J.; Catapano, A.L.; Descamps, O.S.; Fisher, E.; Kovanen, P.T.; Kuivenhoven, J.A.; et al. Triglyceride-rich lipoproteins and high-density lipoprotein cholesterol in patients at high risk of cardiovascular disease: Evidence and guidance for management. Eur. Heart J. 2011, 32, 1345–1361. [Google Scholar] [CrossRef]
  16. Ginsberg, H.N.; Packard, C.J.; Chapman, M.J.; Boren, J.; Aguilar-Salinas, C.A.; Averna, M.; Ference, B.A.; Gaudet, D.; Hegele, R.A.; Kersten, S.; et al. Triglyceride-rich lipoproteins and their remnants: Metabolic insights, role in atherosclerotic cardiovascular disease, and emerging therapeutic strategies-a consensus statement from the European Atherosclerosis Society. Eur. Heart J. 2021, 42, 4791–4806. [Google Scholar] [CrossRef] [PubMed]
  17. Goldberg, I.J. Lipoprotein lipase and lipolysis: Central roles in lipoprotein metabolism and atherogenesis. J. Lipid Res. 1996, 37, 693–707. [Google Scholar] [CrossRef]
  18. Gofman, J.W.; Lindgren, F. The role of lipids and lipoproteins in atherosclerosis. Science 1950, 111, 166–171. [Google Scholar] [CrossRef]
  19. Gofman, J.W.; Jones, H.B.; Lyon, T.P.; Lindgren, F.; Strisower, B.; Colman, D.; Herring, V. Blood lipids and human atherosclerosis. Circulation 1952, 5, 119–134. [Google Scholar] [CrossRef]
  20. Goldstein, J.L.; Brown, M.S. A century of cholesterol and coronaries: From plaques to genes to statins. Cell 2015, 161, 161–172. [Google Scholar] [CrossRef]
  21. Goldstein, J.L.; Brown, M.S. The LDL receptor. Arter. Thromb. Vasc. Biol. 2009, 29, 431–438. [Google Scholar] [CrossRef]
  22. Berndsen, Z.T.; Cassidy, C.K. The structure of apolipoprotein B100 from human low-density lipoprotein. Nature 2025, 638, 836–843. [Google Scholar] [CrossRef] [PubMed]
  23. Reimund, M.; Dearborn, A.D.; Graziano, G.; Lei, H.; Ciancone, A.M.; Kumar, A.; Holewinski, R.; Neufeld, E.B.; O’Reilly, F.J.; Remaley, A.T.; et al. Structure of apolipoprotein B100 bound to the low-density lipoprotein receptor. Nature 2025, 638, 829–835. [Google Scholar] [CrossRef]
  24. Brown, M.S.; Goldstein, J.L. A receptor-mediated pathway for cholesterol homeostasis. Science 1986, 232, 34–47. [Google Scholar] [CrossRef] [PubMed]
  25. Davidson, W.S. The structure of bad cholesterol comes into focus. Nature 2025, 638, 618–620. [Google Scholar] [CrossRef] [PubMed]
  26. Powers, H.R.; Sahoo, D. SR-B1’s Next Top Model: Structural Perspectives on the Functions of the HDL Receptor. Curr. Atheroscler. Rep. 2022, 24, 277–288. [Google Scholar] [CrossRef]
  27. Linton, M.F.; Yancey, P.G.; Tao, H.; Davies, S.S. HDL Function and Atherosclerosis: Reactive Dicarbonyls as Promising Targets of Therapy. Circ. Res. 2023, 132, 1521–1545. [Google Scholar] [CrossRef]
  28. Ouimet, M.; Barrett, T.J.; Fisher, E.A. HDL and Reverse Cholesterol Transport. Circ. Res. 2019, 124, 1505–1518. [Google Scholar] [CrossRef]
  29. Bae, H.; Jung, S.; Le, J.; Tamburini, I.; Kim, J.; Wang, E.; Song, W.S.; Choi, W.; Jang, K.H.; Kang, T.; et al. Cross-organ metabolite production and consumption in healthy and atherogenic conditions. Cell 2025, 188, 4441–4455.e16. [Google Scholar] [CrossRef]
  30. Sturm, A.C.; Knowles, J.W.; Gidding, S.S.; Ahmad, Z.S.; Ahmed, C.D.; Ballantyne, C.M.; Baum, S.J.; Bourbon, M.; Carrie, A.; Cuchel, M.; et al. Clinical Genetic Testing for Familial Hypercholesterolemia: JACC Scientific Expert Panel. J. Am. Coll. Cardiol. 2018, 72, 662–680. [Google Scholar] [CrossRef]
  31. McGowan, M.P.; Hosseini Dehkordi, S.H.; Moriarty, P.M.; Duell, P.B. Diagnosis and Treatment of Heterozygous Familial Hypercholesterolemia. J. Am. Heart Assoc. 2019, 8, e013225. [Google Scholar] [CrossRef]
  32. Lee, Y.; Siddiqui, W.J. Cholesterol Levels. In StatPearls; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
  33. Farzam, K.; Zubair, M.; Senthilkumaran, S. Lipoprotein A. In StatPearls; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
  34. Koschinsky, M.L.; Bajaj, A.; Boffa, M.B.; Dixon, D.L.; Ferdinand, K.C.; Gidding, S.S.; Gill, E.A.; Jacobson, T.A.; Michos, E.D.; Safarova, M.S.; et al. A focused update to the 2019 NLA scientific statement on use of lipoprotein(a) in clinical practice. J Clin Lipidol 2024, 18, e308–e319. [Google Scholar] [CrossRef] [PubMed]
  35. Tsimikas, S.; Fazio, S.; Ferdinand, K.C.; Ginsberg, H.N.; Koschinsky, M.L.; Marcovina, S.M.; Moriarty, P.M.; Rader, D.J.; Remaley, A.T.; Reyes-Soffer, G.; et al. NHLBI Working Group Recommendations to Reduce Lipoprotein(a)-Mediated Risk of Cardiovascular Disease and Aortic Stenosis. J. Am. Coll. Cardiol. 2018, 71, 177–192. [Google Scholar] [CrossRef] [PubMed]
  36. Madsen, C.M.; Kamstrup, P.R.; Langsted, A.; Varbo, A.; Nordestgaard, B.G. Lipoprotein(a)-Lowering by 50 mg/dL (105 nmol/L) May Be Needed to Reduce Cardiovascular Disease 20% in Secondary Prevention: A Population-Based Study. Arter. Thromb. Vasc. Biol. 2020, 40, 255–266. [Google Scholar] [CrossRef]
  37. Ross, R.; Glomset, J.A. The pathogenesis of atherosclerosis (first of two parts). N. Engl. J. Med. 1976, 295, 369–377. [Google Scholar] [CrossRef]
  38. Ross, R.; Glomset, J.A. The pathogenesis of atherosclerosis (second of two parts). N. Engl. J. Med. 1976, 295, 420–425. [Google Scholar] [CrossRef]
  39. Ference, B.A.; Braunwald, E.; Catapano, A.L. The LDL cumulative exposure hypothesis: Evidence and practical applications. Nat. Rev. Cardiol. 2024, 21, 701–716. [Google Scholar] [CrossRef] [PubMed]
  40. Williams, K.J.; Tabas, I. The response-to-retention hypothesis of early atherogenesis. Arter. Thromb. Vasc. Biol. 1995, 15, 551–561. [Google Scholar] [CrossRef] [PubMed]
  41. Steinberg, D. Atherogenesis in perspective: Hypercholesterolemia and inflammation as partners in crime. Nat. Med. 2002, 8, 1211–1217. [Google Scholar] [CrossRef]
  42. Cheng, C.K.; Wang, N.; Wang, L.; Huang, Y. Biophysical and Biochemical Roles of Shear Stress on Endothelium: A Revisit and New Insights. Circ. Res. 2025, 136, 752–772. [Google Scholar] [CrossRef] [PubMed]
  43. Souilhol, C.; Serbanovic-Canic, J.; Fragiadaki, M.; Chico, T.J.; Ridger, V.; Roddie, H.; Evans, P.C. Endothelial responses to shear stress in atherosclerosis: A novel role for developmental genes. Nat. Rev. Cardiol. 2020, 17, 52–63. [Google Scholar] [CrossRef]
  44. Botts, S.R.; Fish, J.E.; Howe, K.L. Dysfunctional Vascular Endothelium as a Driver of Atherosclerosis: Emerging Insights Into Pathogenesis and Treatment. Front. Pharmacol. 2021, 12, 787541. [Google Scholar] [CrossRef]
  45. Gimbrone, M.A., Jr.; Garcia-Cardena, G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016, 118, 620–636. [Google Scholar] [CrossRef]
  46. Souilhol, C.; Harmsen, M.C.; Evans, P.C.; Krenning, G. Endothelial-mesenchymal transition in atherosclerosis. Cardiovasc. Res. 2018, 114, 565–577. [Google Scholar] [CrossRef]
  47. De Caterina, R.; Libby, P.; Peng, H.B.; Thannickal, V.J.; Rajavashisth, T.B.; Gimbrone, M.A., Jr.; Shin, W.S.; Liao, J.K. Nitric oxide decreases cytokine-induced endothelial activation. Nitric oxide selectively reduces endothelial expression of adhesion molecules and proinflammatory cytokines. J. Clin. Investig. 1995, 96, 60–68. [Google Scholar] [CrossRef]
  48. Marshall, H.E.; Stamler, J.S. Nitrosative stress-induced apoptosis through inhibition of NF-kappa B. J. Biol. Chem. 2002, 277, 34223–34228. [Google Scholar] [CrossRef]
  49. Matthews, J.R.; Botting, C.H.; Panico, M.; Morris, H.R.; Hay, R.T. Inhibition of NF-kappaB DNA binding by nitric oxide. Nucleic Acids Res. 1996, 24, 2236–2242. [Google Scholar] [CrossRef] [PubMed]
  50. Reynaert, N.L.; Ckless, K.; Korn, S.H.; Vos, N.; Guala, A.S.; Wouters, E.F.; van der Vliet, A.; Janssen-Heininger, Y.M. Nitric oxide represses inhibitory kappaB kinase through S-nitrosylation. Proc. Natl. Acad. Sci. USA 2004, 101, 8945–8950. [Google Scholar] [CrossRef] [PubMed]
  51. Mundi, S.; Massaro, M.; Scoditti, E.; Carluccio, M.A.; van Hinsbergh, V.W.M.; Iruela-Arispe, M.L.; De Caterina, R. Endothelial permeability, LDL deposition, and cardiovascular risk factors-a review. Cardiovasc. Res. 2018, 114, 35–52. [Google Scholar] [CrossRef]
  52. Hirata, K.; Akita, H.; Yokoyama, M. Oxidized low density lipoprotein inhibits bradykinin-induced phosphoinositide hydrolysis in cultured bovine aortic endothelial cells. FEBS Lett. 1991, 287, 181–184. [Google Scholar] [CrossRef] [PubMed]
  53. Inoue, N.; Hirata, K.; Yamada, M.; Hamamori, Y.; Matsuda, Y.; Akita, H.; Yokoyama, M. Lysophosphatidylcholine inhibits bradykinin-induced phosphoinositide hydrolysis and calcium transients in cultured bovine aortic endothelial cells. Circ. Res. 1992, 71, 1410–1421. [Google Scholar] [CrossRef]
  54. Miwa, Y.; Hirata, K.; Kawashima, S.; Akita, H.; Yokoyama, M. Lysophosphatidylcholine inhibits receptor-mediated Ca2+ mobilization in intact endothelial cells of rabbit aorta. Arter. Thromb. Vasc. Biol. 1997, 17, 1561–1567. [Google Scholar] [CrossRef]
  55. Feron, O.; Dessy, C.; Moniotte, S.; Desager, J.P.; Balligand, J.L. Hypercholesterolemia decreases nitric oxide production by promoting the interaction of caveolin and endothelial nitric oxide synthase. J. Clin. Investig. 1999, 103, 897–905. [Google Scholar] [CrossRef]
  56. Nielsen, L.B. Transfer of low density lipoprotein into the arterial wall and risk of atherosclerosis. Atherosclerosis 1996, 123, 1–15. [Google Scholar] [CrossRef]
  57. Bolanle, I.O.; de Liedekerke Beaufort, G.C.; Weinberg, P.D. Transcytosis of LDL Across Arterial Endothelium: Mechanisms and Therapeutic Targets. Arter. Thromb. Vasc. Biol. 2025, 45, 468–480. [Google Scholar] [CrossRef]
  58. Boren, J.; Chapman, M.J.; Krauss, R.M.; Packard, C.J.; Bentzon, J.F.; Binder, C.J.; Daemen, M.J.; Demer, L.L.; Hegele, R.A.; Nicholls, S.J.; et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease: Pathophysiological, genetic, and therapeutic insights: A consensus statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2020, 41, 2313–2330. [Google Scholar] [CrossRef]
  59. Huang, L.; Chambliss, K.L.; Gao, X.; Yuhanna, I.S.; Behling-Kelly, E.; Bergaya, S.; Ahmed, M.; Michaely, P.; Luby-Phelps, K.; Darehshouri, A.; et al. SR-B1 drives endothelial cell LDL transcytosis via DOCK4 to promote atherosclerosis. Nature 2019, 569, 565–569. [Google Scholar] [CrossRef]
  60. Fung, K.Y.Y.; Ho, T.W.W.; Xu, Z.; Neculai, D.; Beauchemin, C.A.A.; Lee, W.L.; Fairn, G.D. Apolipoprotein A1 and high-density lipoprotein limit low-density lipoprotein transcytosis by binding SR-B1. J. Lipid Res. 2024, 65, 100530. [Google Scholar] [CrossRef] [PubMed]
  61. Nishi, K.; Itabe, H.; Uno, M.; Kitazato, K.T.; Horiguchi, H.; Shinno, K.; Nagahiro, S. Oxidized LDL in carotid plaques and plasma associates with plaque instability. Arter. Thromb. Vasc. Biol. 2002, 22, 1649–1654. [Google Scholar] [CrossRef]
  62. Ohlsson, B.G.; Englund, M.C.; Karlsson, A.L.; Knutsen, E.; Erixon, C.; Skribeck, H.; Liu, Y.; Bondjers, G.; Wiklund, O. Oxidized low density lipoprotein inhibits lipopolysaccharide-induced binding of nuclear factor-kappaB to DNA and the subsequent expression of tumor necrosis factor-alpha and interleukin-1beta in macrophages. J. Clin. Investig. 1996, 98, 78–89. [Google Scholar] [CrossRef]
  63. Li, D.; Mehta, J.L. Upregulation of endothelial receptor for oxidized LDL (LOX-1) by oxidized LDL and implications in apoptosis of human coronary artery endothelial cells: Evidence from use of antisense LOX-1 mRNA and chemical inhibitors. Arter. Thromb. Vasc. Biol. 2000, 20, 1116–1122. [Google Scholar] [CrossRef]
  64. Sawamura, T.; Kume, N.; Aoyama, T.; Moriwaki, H.; Hoshikawa, H.; Aiba, Y.; Tanaka, T.; Miwa, S.; Katsura, Y.; Kita, T.; et al. An endothelial receptor for oxidized low-density lipoprotein. Nature 1997, 386, 73–77. [Google Scholar] [CrossRef] [PubMed]
  65. Mehta, J.L.; Li, D.Y. Identification and autoregulation of receptor for OX-LDL in cultured human coronary artery endothelial cells. Biochem. Biophys. Res. Commun. 1998, 248, 511–514. [Google Scholar] [CrossRef] [PubMed]
  66. Cominacini, L.; Pasini, A.F.; Garbin, U.; Davoli, A.; Tosetti, M.L.; Campagnola, M.; Rigoni, A.; Pastorino, A.M.; Lo Cascio, V.; Sawamura, T. Oxidized low density lipoprotein (ox-LDL) binding to ox-LDL receptor-1 in endothelial cells induces the activation of NF-kappaB through an increased production of intracellular reactive oxygen species. J. Biol. Chem. 2000, 275, 12633–12638. [Google Scholar] [CrossRef]
  67. Galkina, E.; Ley, K. Vascular adhesion molecules in atherosclerosis. Arter. Thromb. Vasc. Biol. 2007, 27, 2292–2301. [Google Scholar] [CrossRef]
  68. Johnson-Tidey, R.R.; McGregor, J.L.; Taylor, P.R.; Poston, R.N. Increase in the adhesion molecule P-selectin in endothelium overlying atherosclerotic plaques. Coexpression with intercellular adhesion molecule-1. Am. J. Pathol. 1994, 144, 952–961. [Google Scholar] [PubMed]
  69. Collins, R.G.; Velji, R.; Guevara, N.V.; Hicks, M.J.; Chan, L.; Beaudet, A.L. P-Selectin or intercellular adhesion molecule (ICAM)-1 deficiency substantially protects against atherosclerosis in apolipoprotein E-deficient mice. J. Exp. Med. 2000, 191, 189–194. [Google Scholar] [CrossRef] [PubMed]
  70. Dong, Z.M.; Brown, A.A.; Wagner, D.D. Prominent role of P-selectin in the development of advanced atherosclerosis in ApoE-deficient mice. Circulation 2000, 101, 2290–2295. [Google Scholar] [CrossRef]
  71. Dong, Z.M.; Chapman, S.M.; Brown, A.A.; Frenette, P.S.; Hynes, R.O.; Wagner, D.D. The combined role of P- and E-selectins in atherosclerosis. J. Clin. Investig. 1998, 102, 145–152. [Google Scholar] [CrossRef]
  72. Davies, M.J.; Gordon, J.L.; Gearing, A.J.; Pigott, R.; Woolf, N.; Katz, D.; Kyriakopoulos, A. The expression of the adhesion molecules ICAM-1, VCAM-1, PECAM, and E-selectin in human atherosclerosis. J. Pathol. 1993, 171, 223–229. [Google Scholar] [CrossRef]
  73. Gerhardt, T.; Ley, K. Monocyte trafficking across the vessel wall. Cardiovasc. Res. 2015, 107, 321–330. [Google Scholar] [CrossRef]
  74. Cybulsky, M.I.; Gimbrone, M.A., Jr. Endothelial expression of a mononuclear leukocyte adhesion molecule during atherogenesis. Science 1991, 251, 788–791. [Google Scholar] [CrossRef]
  75. O’Brien, K.D.; Allen, M.D.; McDonald, T.O.; Chait, A.; Harlan, J.M.; Fishbein, D.; McCarty, J.; Ferguson, M.; Hudkins, K.; Benjamin, C.D.; et al. Vascular cell adhesion molecule-1 is expressed in human coronary atherosclerotic plaques. Implications for the mode of progression of advanced coronary atherosclerosis. J. Clin. Investig. 1993, 92, 945–951. [Google Scholar] [CrossRef] [PubMed]
  76. Nageh, M.F.; Sandberg, E.T.; Marotti, K.R.; Lin, A.H.; Melchior, E.P.; Bullard, D.C.; Beaudet, A.L. Deficiency of inflammatory cell adhesion molecules protects against atherosclerosis in mice. Arter. Thromb. Vasc. Biol. 1997, 17, 1517–1520. [Google Scholar] [CrossRef]
  77. Kitagawa, K.; Matsumoto, M.; Sasaki, T.; Hashimoto, H.; Kuwabara, K.; Ohtsuki, T.; Hori, M. Involvement of ICAM-1 in the progression of atherosclerosis in APOE-knockout mice. Atherosclerosis 2002, 160, 305–310. [Google Scholar] [CrossRef] [PubMed]
  78. Li, H.; Cybulsky, M.I.; Gimbrone, M.A., Jr.; Libby, P. An atherogenic diet rapidly induces VCAM-1, a cytokine-regulatable mononuclear leukocyte adhesion molecule, in rabbit aortic endothelium. Arter. Thromb. 1993, 13, 197–204. [Google Scholar] [CrossRef]
  79. Cybulsky, M.I.; Iiyama, K.; Li, H.; Zhu, S.; Chen, M.; Iiyama, M.; Davis, V.; Gutierrez-Ramos, J.C.; Connelly, P.W.; Milstone, D.S. A major role for VCAM-1, but not ICAM-1, in early atherosclerosis. J. Clin. Investig. 2001, 107, 1255–1262. [Google Scholar] [CrossRef]
  80. Zarbock, A.; Ley, K.; McEver, R.P.; Hidalgo, A. Leukocyte ligands for endothelial selectins: Specialized glycoconjugates that mediate rolling and signaling under flow. Blood 2011, 118, 6743–6751. [Google Scholar] [CrossRef]
  81. Li, D.; Chen, H.; Romeo, F.; Sawamura, T.; Saldeen, T.; Mehta, J.L. Statins modulate oxidized low-density lipoprotein-mediated adhesion molecule expression in human coronary artery endothelial cells: Role of LOX-1. J. Pharmacol. Exp. Ther. 2002, 302, 601–605. [Google Scholar] [CrossRef]
  82. Tacke, F.; Alvarez, D.; Kaplan, T.J.; Jakubzick, C.; Spanbroek, R.; Llodra, J.; Garin, A.; Liu, J.; Mack, M.; van Rooijen, N.; et al. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J. Clin. Investig. 2007, 117, 185–194. [Google Scholar] [CrossRef]
  83. Barron, H.V.; Cannon, C.P.; Murphy, S.A.; Braunwald, E.; Gibson, C.M. Association between white blood cell count, epicardial blood flow, myocardial perfusion, and clinical outcomes in the setting of acute myocardial infarction: A thrombolysis in myocardial infarction 10 substudy. Circulation 2000, 102, 2329–2334. [Google Scholar] [CrossRef]
  84. Stewart, R.A.; White, H.D.; Kirby, A.C.; Heritier, S.R.; Simes, R.J.; Nestel, P.J.; West, M.J.; Colquhoun, D.M.; Tonkin, A.M.; Long-Term Intervention With Pravastatin in Ischemic Disease Study Investigator. White blood cell count predicts reduction in coronary heart disease mortality with pravastatin. Circulation 2005, 111, 1756–1762. [Google Scholar] [CrossRef] [PubMed]
  85. Tani, S.; Nagao, K.; Anazawa, T.; Kawamata, H.; Furuya, S.; Takahashi, H.; Iida, K.; Matsumoto, M.; Washio, T.; Kumabe, N.; et al. Association of leukocyte subtype counts with coronary atherosclerotic regression following pravastatin treatment. Am. J. Cardiol. 2009, 104, 464–469. [Google Scholar] [CrossRef] [PubMed]
  86. Ley, K.; Miller, Y.I.; Hedrick, C.C. Monocyte and macrophage dynamics during atherogenesis. Arter. Thromb. Vasc. Biol. 2011, 31, 1506–1516. [Google Scholar] [CrossRef]
  87. Hilgendorf, I.; Swirski, F.K.; Robbins, C.S. Monocyte fate in atherosclerosis. Arter. Thromb. Vasc. Biol. 2015, 35, 272–279. [Google Scholar] [CrossRef]
  88. Geissmann, F.; Jung, S.; Littman, D.R. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 2003, 19, 71–82. [Google Scholar] [CrossRef] [PubMed]
  89. Mildner, A.; Kim, K.W.; Yona, S. Unravelling monocyte functions: From the guardians of health to the regulators of disease. Discov. Immunol. 2024, 3, kyae014. [Google Scholar] [CrossRef] [PubMed]
  90. Swirski, F.K.; Libby, P.; Aikawa, E.; Alcaide, P.; Luscinskas, F.W.; Weissleder, R.; Pittet, M.J. Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J. Clin. Investig. 2007, 117, 195–205. [Google Scholar] [CrossRef]
  91. Robbins, C.S.; Chudnovskiy, A.; Rauch, P.J.; Figueiredo, J.L.; Iwamoto, Y.; Gorbatov, R.; Etzrodt, M.; Weber, G.F.; Ueno, T.; van Rooijen, N.; et al. Extramedullary hematopoiesis generates Ly-6C(high) monocytes that infiltrate atherosclerotic lesions. Circulation 2012, 125, 364–374. [Google Scholar] [CrossRef]
  92. Emami, H.; Singh, P.; MacNabb, M.; Vucic, E.; Lavender, Z.; Rudd, J.H.; Fayad, Z.A.; Lehrer-Graiwer, J.; Korsgren, M.; Figueroa, A.L.; et al. Splenic metabolic activity predicts risk of future cardiovascular events: Demonstration of a cardiosplenic axis in humans. JACC Cardiovasc. Imaging 2015, 8, 121–130. [Google Scholar] [CrossRef] [PubMed]
  93. Gleissner, C.A.; Leitinger, N.; Ley, K. Effects of native and modified low-density lipoproteins on monocyte recruitment in atherosclerosis. Hypertension 2007, 50, 276–283. [Google Scholar] [CrossRef]
  94. Han, K.H.; Han, K.O.; Green, S.R.; Quehenberger, O. Expression of the monocyte chemoattractant protein-1 receptor CCR2 is increased in hypercholesterolemia. Differential effects of plasma lipoproteins on monocyte function. J. Lipid Res. 1999, 40, 1053–1063. [Google Scholar] [CrossRef]
  95. Han, K.H.; Tangirala, R.K.; Green, S.R.; Quehenberger, O. Chemokine receptor CCR2 expression and monocyte chemoattractant protein-1-mediated chemotaxis in human monocytes. A regulatory role for plasma LDL. Arter. Thromb. Vasc. Biol. 1998, 18, 1983–1991. [Google Scholar] [CrossRef]
  96. Cushing, S.D.; Berliner, J.A.; Valente, A.J.; Territo, M.C.; Navab, M.; Parhami, F.; Gerrity, R.; Schwartz, C.J.; Fogelman, A.M. Minimally modified low density lipoprotein induces monocyte chemotactic protein 1 in human endothelial cells and smooth muscle cells. Proc. Natl. Acad. Sci. USA 1990, 87, 5134–5138. [Google Scholar] [CrossRef]
  97. Lee, H.; Shi, W.; Tontonoz, P.; Wang, S.; Subbanagounder, G.; Hedrick, C.C.; Hama, S.; Borromeo, C.; Evans, R.M.; Berliner, J.A.; et al. Role for peroxisome proliferator-activated receptor alpha in oxidized phospholipid-induced synthesis of monocyte chemotactic protein-1 and interleukin-8 by endothelial cells. Circ. Res. 2000, 87, 516–521. [Google Scholar] [CrossRef] [PubMed]
  98. Suriyaphol, P.; Fenske, D.; Zahringer, U.; Han, S.R.; Bhakdi, S.; Husmann, M. Enzymatically modified nonoxidized low-density lipoprotein induces interleukin-8 in human endothelial cells: Role of free fatty acids. Circulation 2002, 106, 2581–2587. [Google Scholar] [CrossRef]
  99. Parhami, F.; Fang, Z.T.; Fogelman, A.M.; Andalibi, A.; Territo, M.C.; Berliner, J.A. Minimally modified low density lipoprotein-induced inflammatory responses in endothelial cells are mediated by cyclic adenosine monophosphate. J. Clin. Investig. 1993, 92, 471–478. [Google Scholar] [CrossRef]
  100. Gerszten, R.E.; Garcia-Zepeda, E.A.; Lim, Y.C.; Yoshida, M.; Ding, H.A.; Gimbrone, M.A., Jr.; Luster, A.D.; Luscinskas, F.W.; Rosenzweig, A. MCP-1 and IL-8 trigger firm adhesion of monocytes to vascular endothelium under flow conditions. Nature 1999, 398, 718–723. [Google Scholar] [CrossRef]
  101. Gu, L.; Okada, Y.; Clinton, S.K.; Gerard, C.; Sukhova, G.K.; Libby, P.; Rollins, B.J. Absence of monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor-deficient mice. Mol. Cell 1998, 2, 275–281. [Google Scholar] [CrossRef]
  102. Boring, L.; Gosling, J.; Cleary, M.; Charo, I.F. Decreased lesion formation in CCR2-/- mice reveals a role for chemokines in the initiation of atherosclerosis. Nature 1998, 394, 894–897. [Google Scholar] [CrossRef]
  103. Boisvert, W.A.; Santiago, R.; Curtiss, L.K.; Terkeltaub, R.A. A leukocyte homologue of the IL-8 receptor CXCR-2 mediates the accumulation of macrophages in atherosclerotic lesions of LDL receptor-deficient mice. J. Clin. Investig. 1998, 101, 353–363. [Google Scholar] [CrossRef]
  104. Moore, K.J.; Tabas, I. Macrophages in the pathogenesis of atherosclerosis. Cell 2011, 145, 341–355. [Google Scholar] [CrossRef] [PubMed]
  105. Smith, J.D.; Trogan, E.; Ginsberg, M.; Grigaux, C.; Tian, J.; Miyata, M. Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proc. Natl. Acad. Sci. USA 1995, 92, 8264–8268. [Google Scholar] [CrossRef] [PubMed]
  106. Qiao, J.H.; Tripathi, J.; Mishra, N.K.; Cai, Y.; Tripathi, S.; Wang, X.P.; Imes, S.; Fishbein, M.C.; Clinton, S.K.; Libby, P.; et al. Role of macrophage colony-stimulating factor in atherosclerosis: Studies of osteopetrotic mice. Am. J. Pathol. 1997, 150, 1687–1699. [Google Scholar]
  107. Rajavashisth, T.B.; Andalibi, A.; Territo, M.C.; Berliner, J.A.; Navab, M.; Fogelman, A.M.; Lusis, A.J. Induction of endothelial cell expression of granulocyte and macrophage colony-stimulating factors by modified low-density lipoproteins. Nature 1990, 344, 254–257. [Google Scholar] [CrossRef]
  108. Sinha, S.K.; Miikeda, A.; Fouladian, Z.; Mehrabian, M.; Edillor, C.; Shih, D.; Zhou, Z.; Paul, M.K.; Charugundla, S.; Davis, R.C.; et al. Local M-CSF (Macrophage Colony-Stimulating Factor) Expression Regulates Macrophage Proliferation and Apoptosis in Atherosclerosis. Arter. Thromb. Vasc. Biol. 2021, 41, 220–233. [Google Scholar] [CrossRef]
  109. Bjorkegren, J.L.M.; Lusis, A.J. Atherosclerosis: Recent developments. Cell 2022, 185, 1630–1645. [Google Scholar] [CrossRef] [PubMed]
  110. Yoshida, H.; Quehenberger, O.; Kondratenko, N.; Green, S.; Steinberg, D. Minimally oxidized low-density lipoprotein increases expression of scavenger receptor A, CD36, and macrosialin in resident mouse peritoneal macrophages. Arter. Thromb. Vasc. Biol. 1998, 18, 794–802. [Google Scholar] [CrossRef] [PubMed]
  111. Avogaro, P.; Bon, G.B.; Cazzolato, G. Presence of a modified low density lipoprotein in humans. Arteriosclerosis 1988, 8, 79–87. [Google Scholar] [CrossRef]
  112. Boren, J.; Olin, K.; Lee, I.; Chait, A.; Wight, T.N.; Innerarity, T.L. Identification of the principal proteoglycan-binding site in LDL. A single-point mutation in apo-B100 severely affects proteoglycan interaction without affecting LDL receptor binding. J. Clin. Investig. 1998, 101, 2658–2664. [Google Scholar] [CrossRef] [PubMed]
  113. Kaplan, M.; Aviram, M. Retention of oxidized LDL by extracellular matrix proteoglycans leads to its uptake by macrophages: An alternative approach to study lipoproteins cellular uptake. Arter. Thromb. Vasc. Biol. 2001, 21, 386–393. [Google Scholar] [CrossRef]
  114. Stewart, C.R.; Stuart, L.M.; Wilkinson, K.; van Gils, J.M.; Deng, J.; Halle, A.; Rayner, K.J.; Boyer, L.; Zhong, R.; Frazier, W.A.; et al. CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat. Immunol. 2010, 11, 155–161. [Google Scholar] [CrossRef]
  115. Sheedy, F.J.; Grebe, A.; Rayner, K.J.; Kalantari, P.; Ramkhelawon, B.; Carpenter, S.B.; Becker, C.E.; Ediriweera, H.N.; Mullick, A.E.; Golenbock, D.T.; et al. CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat. Immunol. 2013, 14, 812–820. [Google Scholar] [CrossRef]
  116. Kodama, T.; Reddy, P.; Kishimoto, C.; Krieger, M. Purification and characterization of a bovine acetyl low density lipoprotein receptor. Proc. Natl. Acad. Sci. USA 1988, 85, 9238–9242. [Google Scholar] [CrossRef]
  117. Kodama, T.; Freeman, M.; Rohrer, L.; Zabrecky, J.; Matsudaira, P.; Krieger, M. Type I macrophage scavenger receptor contains alpha-helical and collagen-like coiled coils. Nature 1990, 343, 531–535. [Google Scholar] [CrossRef]
  118. Seimon, T.A.; Nadolski, M.J.; Liao, X.; Magallon, J.; Nguyen, M.; Feric, N.T.; Koschinsky, M.L.; Harkewicz, R.; Witztum, J.L.; Tsimikas, S.; et al. Atherogenic lipids and lipoproteins trigger CD36-TLR2-dependent apoptosis in macrophages undergoing endoplasmic reticulum stress. Cell Metab. 2010, 12, 467–482. [Google Scholar] [CrossRef]
  119. Tian, K.; Xu, Y.; Sahebkar, A.; Xu, S. CD36 in Atherosclerosis: Pathophysiological Mechanisms and Therapeutic Implications. Curr. Atheroscler. Rep. 2020, 22, 59. [Google Scholar] [CrossRef]
  120. Febbraio, M.; Hajjar, D.P.; Silverstein, R.L. CD36: A class B scavenger receptor involved in angiogenesis, atherosclerosis, inflammation, and lipid metabolism. J. Clin. Investig. 2001, 108, 785–791. [Google Scholar] [CrossRef] [PubMed]
  121. de Winther, M.P.; van Dijk, K.W.; Havekes, L.M.; Hofker, M.H. Macrophage scavenger receptor class A: A multifunctional receptor in atherosclerosis. Arter. Thromb. Vasc. Biol. 2000, 20, 290–297. [Google Scholar] [CrossRef] [PubMed]
  122. Schaeffer, D.F.; Riazy, M.; Parhar, K.S.; Chen, J.H.; Duronio, V.; Sawamura, T.; Steinbrecher, U.P. LOX-1 augments oxLDL uptake by lysoPC-stimulated murine macrophages but is not required for oxLDL clearance from plasma. J. Lipid Res. 2009, 50, 1676–1684. [Google Scholar] [CrossRef] [PubMed]
  123. Talle, M.A.; Rao, P.E.; Westberg, E.; Allegar, N.; Makowski, M.; Mittler, R.S.; Goldstein, G. Patterns of antigenic expression on human monocytes as defined by monoclonal antibodies. Cell Immunol. 1983, 78, 83–99. [Google Scholar] [CrossRef]
  124. Rahaman, S.O.; Lennon, D.J.; Febbraio, M.; Podrez, E.A.; Hazen, S.L.; Silverstein, R.L. A CD36-dependent signaling cascade is necessary for macrophage foam cell formation. Cell Metab. 2006, 4, 211–221. [Google Scholar] [CrossRef]
  125. Febbraio, M.; Podrez, E.A.; Smith, J.D.; Hajjar, D.P.; Hazen, S.L.; Hoff, H.F.; Sharma, K.; Silverstein, R.L. Targeted disruption of the class B scavenger receptor CD36 protects against atherosclerotic lesion development in mice. J. Clin. Investig. 2000, 105, 1049–1056. [Google Scholar] [CrossRef]
  126. Moore, K.J.; Kunjathoor, V.V.; Koehn, S.L.; Manning, J.J.; Tseng, A.A.; Silver, J.M.; McKee, M.; Freeman, M.W. Loss of receptor-mediated lipid uptake via scavenger receptor A or CD36 pathways does not ameliorate atherosclerosis in hyperlipidemic mice. J. Clin. Investig. 2005, 115, 2192–2201. [Google Scholar] [CrossRef]
  127. Manning-Tobin, J.J.; Moore, K.J.; Seimon, T.A.; Bell, S.A.; Sharuk, M.; Alvarez-Leite, J.I.; de Winther, M.P.; Tabas, I.; Freeman, M.W. Loss of SR-A and CD36 activity reduces atherosclerotic lesion complexity without abrogating foam cell formation in hyperlipidemic mice. Arter. Thromb. Vasc. Biol. 2009, 29, 19–26. [Google Scholar] [CrossRef]
  128. Singh, R.K.; Haka, A.S.; Asmal, A.; Barbosa-Lorenzi, V.C.; Grosheva, I.; Chin, H.F.; Xiong, Y.; Hla, T.; Maxfield, F.R. TLR4 (Toll-Like Receptor 4)-Dependent Signaling Drives Extracellular Catabolism of LDL (Low-Density Lipoprotein) Aggregates. Arter. Thromb. Vasc. Biol. 2020, 40, 86–102. [Google Scholar] [CrossRef] [PubMed]
  129. Lin, H.P.; Singla, B.; Ahn, W.; Ghoshal, P.; Blahove, M.; Cherian-Shaw, M.; Chen, A.; Haller, A.; Hui, D.Y.; Dong, K.; et al. Receptor-independent fluid-phase macropinocytosis promotes arterial foam cell formation and atherosclerosis. Sci. Transl. Med. 2022, 14, eadd2376. [Google Scholar] [CrossRef]
  130. Tabas, I. Cholesterol and phospholipid metabolism in macrophages. Biochim. Biophys. Acta 2000, 1529, 164–174. [Google Scholar] [CrossRef] [PubMed]
  131. Ouimet, M.; Franklin, V.; Mak, E.; Liao, X.; Tabas, I.; Marcel, Y.L. Autophagy regulates cholesterol efflux from macrophage foam cells via lysosomal acid lipase. Cell Metab. 2011, 13, 655–667. [Google Scholar] [CrossRef] [PubMed]
  132. Remaley, A.T.; Stonik, J.A.; Demosky, S.J.; Neufeld, E.B.; Bocharov, A.V.; Vishnyakova, T.G.; Eggerman, T.L.; Patterson, A.P.; Duverger, N.J.; Santamarina-Fojo, S.; et al. Apolipoprotein specificity for lipid efflux by the human ABCAI transporter. Biochem. Biophys. Res. Commun. 2001, 280, 818–823. [Google Scholar] [CrossRef]
  133. Kennedy, M.A.; Barrera, G.C.; Nakamura, K.; Baldan, A.; Tarr, P.; Fishbein, M.C.; Frank, J.; Francone, O.L.; Edwards, P.A. ABCG1 has a critical role in mediating cholesterol efflux to HDL and preventing cellular lipid accumulation. Cell Metab. 2005, 1, 121–131. [Google Scholar] [CrossRef]
  134. Brown, A.J.; Jessup, W. Oxysterols and atherosclerosis. Atherosclerosis 1999, 142, 1–28. [Google Scholar] [CrossRef]
  135. Janowski, B.A.; Willy, P.J.; Devi, T.R.; Falck, J.R.; Mangelsdorf, D.J. An oxysterol signalling pathway mediated by the nuclear receptor LXR alpha. Nature 1996, 383, 728–731. [Google Scholar] [CrossRef]
  136. Liu, Y.; Wei, Z.; Ma, X.; Yang, X.; Chen, Y.; Sun, L.; Ma, C.; Miao, Q.R.; Hajjar, D.P.; Han, J.; et al. 25-Hydroxycholesterol activates the expression of cholesterol 25-hydroxylase in an LXR-dependent mechanism. J. Lipid Res. 2018, 59, 439–451. [Google Scholar] [CrossRef]
  137. Lehmann, J.M.; Kliewer, S.A.; Moore, L.B.; Smith-Oliver, T.A.; Oliver, B.B.; Su, J.L.; Sundseth, S.S.; Winegar, D.A.; Blanchard, D.E.; Spencer, T.A.; et al. Activation of the nuclear receptor LXR by oxysterols defines a new hormone response pathway. J. Biol. Chem. 1997, 272, 3137–3140. [Google Scholar] [CrossRef]
  138. Rasheed, A.; Cummins, C.L. Beyond the Foam Cell: The Role of LXRs in Preventing Atherogenesis. Int. J. Mol. Sci. 2018, 19, 2307. [Google Scholar] [CrossRef] [PubMed]
  139. Joseph, S.B.; Castrillo, A.; Laffitte, B.A.; Mangelsdorf, D.J.; Tontonoz, P. Reciprocal regulation of inflammation and lipid metabolism by liver X receptors. Nat. Med. 2003, 9, 213–219. [Google Scholar] [CrossRef]
  140. Tangirala, R.K.; Bischoff, E.D.; Joseph, S.B.; Wagner, B.L.; Walczak, R.; Laffitte, B.A.; Daige, C.L.; Thomas, D.; Heyman, R.A.; Mangelsdorf, D.J.; et al. Identification of macrophage liver X receptors as inhibitors of atherosclerosis. Proc. Natl. Acad. Sci. USA 2002, 99, 11896–11901. [Google Scholar] [CrossRef]
  141. Alshaikhli, A.; Vaqar, S. Tangier Disease. In StatPearls; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
  142. Oram, J.F. Tangier disease and ABCA1. Biochim. Biophys. Acta 2000, 1529, 321–330. [Google Scholar] [CrossRef]
  143. Ghisletti, S.; Huang, W.; Jepsen, K.; Benner, C.; Hardiman, G.; Rosenfeld, M.G.; Glass, C.K. Cooperative NCoR/SMRT interactions establish a corepressor-based strategy for integration of inflammatory and anti-inflammatory signaling pathways. Genes. Dev. 2009, 23, 681–693. [Google Scholar] [CrossRef] [PubMed]
  144. Thomas, D.G.; Doran, A.C.; Fotakis, P.; Westerterp, M.; Antonson, P.; Jiang, H.; Jiang, X.C.; Gustafsson, J.A.; Tabas, I.; Tall, A.R. LXR Suppresses Inflammatory Gene Expression and Neutrophil Migration through cis-Repression and Cholesterol Efflux. Cell Rep. 2018, 25, 3774–3785.e4. [Google Scholar] [CrossRef] [PubMed]
  145. Zhang, X.; McDonald, J.G.; Aryal, B.; Canfran-Duque, A.; Goldberg, E.L.; Araldi, E.; Ding, W.; Fan, Y.; Thompson, B.M.; Singh, A.K.; et al. Desmosterol suppresses macrophage inflammasome activation and protects against vascular inflammation and atherosclerosis. Proc. Natl. Acad. Sci. USA 2021, 118, e2107682118. [Google Scholar] [CrossRef]
  146. Spann, N.J.; Garmire, L.X.; McDonald, J.G.; Myers, D.S.; Milne, S.B.; Shibata, N.; Reichart, D.; Fox, J.N.; Shaked, I.; Heudobler, D.; et al. Regulated accumulation of desmosterol integrates macrophage lipid metabolism and inflammatory responses. Cell 2012, 151, 138–152. [Google Scholar] [CrossRef]
  147. Allahverdian, S.; Chehroudi, A.C.; McManus, B.M.; Abraham, T.; Francis, G.A. Contribution of intimal smooth muscle cells to cholesterol accumulation and macrophage-like cells in human atherosclerosis. Circulation 2014, 129, 1551–1559. [Google Scholar] [CrossRef]
  148. Feil, S.; Fehrenbacher, B.; Lukowski, R.; Essmann, F.; Schulze-Osthoff, K.; Schaller, M.; Feil, R. Transdifferentiation of vascular smooth muscle cells to macrophage-like cells during atherogenesis. Circ. Res. 2014, 115, 662–667. [Google Scholar] [CrossRef]
  149. Shankman, L.S.; Gomez, D.; Cherepanova, O.A.; Salmon, M.; Alencar, G.F.; Haskins, R.M.; Swiatlowska, P.; Newman, A.A.; Greene, E.S.; Straub, A.C.; et al. KLF4-dependent phenotypic modulation of smooth muscle cells has a key role in atherosclerotic plaque pathogenesis. Nat. Med. 2015, 21, 628–637. [Google Scholar] [CrossRef]
  150. Wang, Y.; Dubland, J.A.; Allahverdian, S.; Asonye, E.; Sahin, B.; Jaw, J.E.; Sin, D.D.; Seidman, M.A.; Leeper, N.J.; Francis, G.A. Smooth Muscle Cells Contribute the Majority of Foam Cells in ApoE (Apolipoprotein E)-Deficient Mouse Atherosclerosis. Arter. Thromb. Vasc. Biol. 2019, 39, 876–887. [Google Scholar] [CrossRef]
  151. Dubland, J.A.; Allahverdian, S.; Besler, K.J.; Ortega, C.; Wang, Y.; Pryma, C.S.; Boukais, K.; Chan, T.; Seidman, M.A.; Francis, G.A. Low LAL (Lysosomal Acid Lipase) Expression by Smooth Muscle Cells Relative to Macrophages as a Mechanism for Arterial Foam Cell Formation. Arter. Thromb. Vasc. Biol. 2021, 41, e354–e368. [Google Scholar] [CrossRef] [PubMed]
  152. Robichaud, S.; Rasheed, A.; Pietrangelo, A.; Doyoung Kim, A.; Boucher, D.M.; Emerton, C.; Vijithakumar, V.; Gharibeh, L.; Fairman, G.; Mak, E.; et al. Autophagy Is Differentially Regulated in Leukocyte and Nonleukocyte Foam Cells During Atherosclerosis. Circ. Res. 2022, 130, 831–847. [Google Scholar] [CrossRef] [PubMed]
  153. Hotamisligil, G.S. Endoplasmic reticulum stress and atherosclerosis. Nat. Med. 2010, 16, 396–399. [Google Scholar] [CrossRef]
  154. Feng, B.; Yao, P.M.; Li, Y.; Devlin, C.M.; Zhang, D.; Harding, H.P.; Sweeney, M.; Rong, J.X.; Kuriakose, G.; Fisher, E.A.; et al. The endoplasmic reticulum is the site of cholesterol-induced cytotoxicity in macrophages. Nat. Cell Biol. 2003, 5, 781–792. [Google Scholar] [CrossRef] [PubMed]
  155. Li, S.; Navia-Pelaez, J.M.; Choi, S.H.; Miller, Y.I. Macrophage inflammarafts in atherosclerosis. Curr. Opin. Lipidol. 2023, 34, 189–195. [Google Scholar] [CrossRef]
  156. Navia-Pelaez, J.M.; Agatisa-Boyle, C.; Choi, S.H.; Sak Kim, Y.; Li, S.; Alekseeva, E.; Weldy, K.; Miller, Y.I. Differential Expression of Inflammarafts in Macrophage Foam Cells and in Nonfoamy Macrophages in Atherosclerotic Lesions-Brief Report. Arter. Thromb. Vasc. Biol. 2023, 43, 323–329. [Google Scholar] [CrossRef]
  157. Canfran-Duque, A.; Rotllan, N.; Zhang, X.; Andres-Blasco, I.; Thompson, B.M.; Sun, J.; Price, N.L.; Fernandez-Fuertes, M.; Fowler, J.W.; Gomez-Coronado, D.; et al. Macrophage-Derived 25-Hydroxycholesterol Promotes Vascular Inflammation, Atherogenesis, and Lesion Remodeling. Circulation 2023, 147, 388–408. [Google Scholar] [CrossRef]
  158. Khan, T.J.; Semenkovich, C.F.; Zayed, M.A. De novo lipid synthesis in cardiovascular tissue and disease. Atherosclerosis 2025, 400, 119066. [Google Scholar] [CrossRef]
  159. Vogel, A.; Brunner, J.S.; Hajto, A.; Sharif, O.; Schabbauer, G. Lipid scavenging macrophages and inflammation. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2022, 1867, 159066. [Google Scholar] [CrossRef]
  160. Rasheed, A.; Rayner, K.J. Macrophage Responses to Environmental Stimuli During Homeostasis and Disease. Endocr. Rev. 2021, 42, 407–435. [Google Scholar] [CrossRef] [PubMed]
  161. Barrett, T.J. Macrophages in Atherosclerosis Regression. Arter. Thromb. Vasc. Biol. 2020, 40, 20–33. [Google Scholar] [CrossRef]
  162. Nomura, M.; Liu, J.; Yu, Z.X.; Yamazaki, T.; Yan, Y.; Kawagishi, H.; Rovira, II; Liu, C.; Wolfgang, M.J.; Mukouyama, Y.S.; et al. Macrophage fatty acid oxidation inhibits atherosclerosis progression. J. Mol. Cell Cardiol. 2019, 127, 270–276. [Google Scholar] [CrossRef]
  163. Kanter, J.E.; Kramer, F.; Barnhart, S.; Averill, M.M.; Vivekanandan-Giri, A.; Vickery, T.; Li, L.O.; Becker, L.; Yuan, W.; Chait, A.; et al. Diabetes promotes an inflammatory macrophage phenotype and atherosclerosis through acyl-CoA synthetase 1. Proc. Natl. Acad. Sci. USA 2012, 109, E715–E724. [Google Scholar] [CrossRef] [PubMed]
  164. Wen, H.; Gris, D.; Lei, Y.; Jha, S.; Zhang, L.; Huang, M.T.; Brickey, W.J.; Ting, J.P. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat. Immunol. 2011, 12, 408–415. [Google Scholar] [CrossRef] [PubMed]
  165. Thorne, R.F.; Ralston, K.J.; de Bock, C.E.; Mhaidat, N.M.; Zhang, X.D.; Boyd, A.W.; Burns, G.F. Palmitoylation of CD36/FAT regulates the rate of its post-transcriptional processing in the endoplasmic reticulum. Biochim. Biophys. Acta 2010, 1803, 1298–1307. [Google Scholar] [CrossRef]
  166. Kumano-Kuramochi, M.; Xie, Q.; Kajiwara, S.; Komba, S.; Minowa, T.; Machida, S. Lectin-like oxidized LDL receptor-1 is palmitoylated and internalizes ligands via caveolae/raft-dependent endocytosis. Biochem. Biophys. Res. Commun. 2013, 434, 594–599. [Google Scholar] [CrossRef]
  167. Dessi, M.; Noce, A.; Bertucci, P.; Manca di Villahermosa, S.; Zenobi, R.; Castagnola, V.; Addessi, E.; Di Daniele, N. Atherosclerosis, dyslipidemia, and inflammation: The significant role of polyunsaturated Fatty acids. ISRN Inflamm. 2013, 2013, 191823. [Google Scholar] [CrossRef]
  168. Wang, B.; Wu, L.; Chen, J.; Dong, L.; Chen, C.; Wen, Z.; Hu, J.; Fleming, I.; Wang, D.W. Metabolism pathways of arachidonic acids: Mechanisms and potential therapeutic targets. Signal Transduct. Target. Ther. 2021, 6, 94. [Google Scholar] [CrossRef]
  169. Yokode, M.; Kita, T.; Kikawa, Y.; Ogorochi, T.; Narumiya, S.; Kawai, C. Stimulated arachidonate metabolism during foam cell transformation of mouse peritoneal macrophages with oxidized low density lipoprotein. J. Clin. Investig. 1988, 81, 720–729. [Google Scholar] [CrossRef]
  170. Zhu, L.; Zhang, Y.; Guo, Z.; Wang, M. Cardiovascular Biology of Prostanoids and Drug Discovery. Arter. Thromb. Vasc. Biol. 2020, 40, 1454–1463. [Google Scholar] [CrossRef]
  171. Shahidi, F.; Ambigaipalan, P. Omega-3 Polyunsaturated Fatty Acids and Their Health Benefits. Annu. Rev. Food Sci. Technol. 2018, 9, 345–381. [Google Scholar] [CrossRef] [PubMed]
  172. Bang, H.O.; Dyerberg, J.; Sinclair, H.M. The composition of the Eskimo food in north western Greenland. Am. J. Clin. Nutr. 1980, 33, 2657–2661. [Google Scholar] [CrossRef] [PubMed]
  173. Davidson, M.; Bulkow, L.R.; Gellin, B.G. Cardiac mortality in Alaska’s indigenous and non-Native residents. Int. J. Epidemiol. 1993, 22, 62–71. [Google Scholar] [CrossRef] [PubMed]
  174. Albert, C.M.; Hennekens, C.H.; O’Donnell, C.J.; Ajani, U.A.; Carey, V.J.; Willett, W.C.; Ruskin, J.N.; Manson, J.E. Fish consumption and risk of sudden cardiac death. JAMA 1998, 279, 23–28. [Google Scholar] [CrossRef]
  175. Yokoyama, M.; Origasa, H.; Matsuzaki, M.; Matsuzawa, Y.; Saito, Y.; Ishikawa, Y.; Oikawa, S.; Sasaki, J.; Hishida, H.; Itakura, H.; et al. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): A randomised open-label, blinded endpoint analysis. Lancet 2007, 369, 1090–1098. [Google Scholar] [CrossRef]
  176. Bhatt, D.L.; Steg, P.G.; Miller, M.; Brinton, E.A.; Jacobson, T.A.; Ketchum, S.B.; Doyle, R.T., Jr.; Juliano, R.A.; Jiao, L.; Granowitz, C.; et al. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N. Engl. J. Med. 2019, 380, 11–22. [Google Scholar] [CrossRef]
  177. Basil, M.C.; Levy, B.D. Specialized pro-resolving mediators: Endogenous regulators of infection and inflammation. Nat. Rev. Immunol. 2016, 16, 51–67. [Google Scholar] [CrossRef]
  178. Stotts, C.; Corrales-Medina, V.F.; Rayner, K.J. Pneumonia-Induced Inflammation, Resolution and Cardiovascular Disease: Causes, Consequences and Clinical Opportunities. Circ. Res. 2023, 132, 751–774. [Google Scholar] [CrossRef]
  179. Ohira, T.; Arita, M.; Omori, K.; Recchiuti, A.; Van Dyke, T.E.; Serhan, C.N. Resolvin E1 receptor activation signals phosphorylation and phagocytosis. J. Biol. Chem. 2010, 285, 3451–3461. [Google Scholar] [CrossRef]
  180. Cash, J.L.; Hart, R.; Russ, A.; Dixon, J.P.; Colledge, W.H.; Doran, J.; Hendrick, A.G.; Carlton, M.B.; Greaves, D.R. Synthetic chemerin-derived peptides suppress inflammation through ChemR23. J. Exp. Med. 2008, 205, 767–775. [Google Scholar] [CrossRef]
  181. Rymut, N.; Heinz, J.; Sadhu, S.; Hosseini, Z.; Riley, C.O.; Marinello, M.; Maloney, J.; MacNamara, K.C.; Spite, M.; Fredman, G. Resolvin D1 promotes efferocytosis in aging by limiting senescent cell-induced MerTK cleavage. FASEB J. 2020, 34, 597–609. [Google Scholar] [CrossRef]
  182. Hosseini, Z.; Marinello, M.; Decker, C.; Sansbury, B.E.; Sadhu, S.; Gerlach, B.D.; Bossardi Ramos, R.; Adam, A.P.; Spite, M.; Fredman, G. Resolvin D1 Enhances Necroptotic Cell Clearance Through Promoting Macrophage Fatty Acid Oxidation and Oxidative Phosphorylation. Arter. Thromb. Vasc. Biol. 2021, 41, 1062–1075. [Google Scholar] [CrossRef]
  183. Arnardottir, H.; Thul, S.; Pawelzik, S.C.; Karadimou, G.; Artiach, G.; Gallina, A.L.; Mysdotter, V.; Carracedo, M.; Tarnawski, L.; Caravaca, A.S.; et al. The resolvin D1 receptor GPR32 transduces inflammation resolution and atheroprotection. J. Clin. Investig. 2021, 131, e142883. [Google Scholar] [CrossRef] [PubMed]
  184. Nordestgaard, B.G. Triglyceride-Rich Lipoproteins and Atherosclerotic Cardiovascular Disease: New Insights From Epidemiology, Genetics, and Biology. Circ. Res. 2016, 118, 547–563. [Google Scholar] [CrossRef] [PubMed]
  185. Bjornson, E.; Adiels, M.; Gummesson, A.; Taskinen, M.R.; Burgess, S.; Packard, C.J.; Boren, J. Quantifying Triglyceride-Rich Lipoprotein Atherogenicity, Associations With Inflammation, and Implications for Risk Assessment Using Non-HDL Cholesterol. J. Am. Coll. Cardiol. 2024, 84, 1328–1338. [Google Scholar] [CrossRef]
  186. Tybjaerg-Hansen, A.; Nordestgaard, B.G.; Christoffersen, M. Triglyceride-rich remnant lipoproteins are more atherogenic than LDL per particle: Is this important? Eur. Heart J. 2023, 44, 4196–4198. [Google Scholar] [CrossRef]
  187. Taskinen, M.R.; Boren, J. Why Is Apolipoprotein CIII Emerging as a Novel Therapeutic Target to Reduce the Burden of Cardiovascular Disease? Curr. Atheroscler. Rep. 2016, 18, 59. [Google Scholar] [CrossRef] [PubMed]
  188. Bornfeldt, K.E. Apolipoprotein C3: Form begets function. J. Lipid Res. 2024, 65, 100475. [Google Scholar] [CrossRef]
  189. Kanter, J.E.; Shao, B.; Kramer, F.; Barnhart, S.; Shimizu-Albergine, M.; Vaisar, T.; Graham, M.J.; Crooke, R.M.; Manuel, C.R.; Haeusler, R.A.; et al. Increased apolipoprotein C3 drives cardiovascular risk in type 1 diabetes. J. Clin. Investig. 2019, 129, 4165–4179. [Google Scholar] [CrossRef]
  190. Qamar, A.; Khetarpal, S.A.; Khera, A.V.; Qasim, A.; Rader, D.J.; Reilly, M.P. Plasma apolipoprotein C-III levels, triglycerides, and coronary artery calcification in type 2 diabetics. Arter. Thromb. Vasc. Biol. 2015, 35, 1880–1888. [Google Scholar] [CrossRef] [PubMed]
  191. Pollin, T.I.; Damcott, C.M.; Shen, H.; Ott, S.H.; Shelton, J.; Horenstein, R.B.; Post, W.; McLenithan, J.C.; Bielak, L.F.; Peyser, P.A.; et al. A null mutation in human APOC3 confers a favorable plasma lipid profile and apparent cardioprotection. Science 2008, 322, 1702–1705. [Google Scholar] [CrossRef] [PubMed]
  192. Wulff, A.B.; Nordestgaard, B.G.; Tybjaerg-Hansen, A. APOC3 Loss-of-Function Mutations, Remnant Cholesterol, Low-Density Lipoprotein Cholesterol, and Cardiovascular Risk: Mediation- and Meta-Analyses of 137 895 Individuals. Arter. Thromb. Vasc. Biol. 2018, 38, 660–668. [Google Scholar] [CrossRef]
  193. Jorgensen, A.B.; Frikke-Schmidt, R.; Nordestgaard, B.G.; Tybjaerg-Hansen, A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N. Engl. J. Med. 2014, 371, 32–41. [Google Scholar] [CrossRef] [PubMed]
  194. Sundaram, M.; Zhong, S.; Bou Khalil, M.; Links, P.H.; Zhao, Y.; Iqbal, J.; Hussain, M.M.; Parks, R.J.; Wang, Y.; Yao, Z. Expression of apolipoprotein C-III in McA-RH7777 cells enhances VLDL assembly and secretion under lipid-rich conditions. J. Lipid Res. 2010, 51, 150–161. [Google Scholar] [CrossRef]
  195. Brown, W.V.; Baginsky, M.L. Inhibition of lipoprotein lipase by an apoprotein of human very low density lipoprotein. Biochem. Biophys. Res. Commun. 1972, 46, 375–382. [Google Scholar] [CrossRef]
  196. Windler, E.; Havel, R.J. Inhibitory effects of C apolipoproteins from rats and humans on the uptake of triglyceride-rich lipoproteins and their remnants by the perfused rat liver. J. Lipid Res. 1985, 26, 556–565. [Google Scholar] [CrossRef]
  197. Mendivil, C.O.; Rimm, E.B.; Furtado, J.; Chiuve, S.E.; Sacks, F.M. Low-density lipoproteins containing apolipoprotein C-III and the risk of coronary heart disease. Circulation 2011, 124, 2065–2072. [Google Scholar] [CrossRef]
  198. Emerging Risk Factors, C.; Erqou, S.; Kaptoge, S.; Perry, P.L.; Di Angelantonio, E.; Thompson, A.; White, I.R.; Marcovina, S.M.; Collins, R.; Thompson, S.G.; et al. Lipoprotein(a) concentration and the risk of coronary heart disease, stroke, and nonvascular mortality. JAMA 2009, 302, 412–423. [Google Scholar] [CrossRef]
  199. Bellomo, T.R.; Bramel, E.E.; Lee, J.; Urbut, S.; Flores, A.; Yu, Z.; Koyama, S.; Truong, B.; Haidermota, S.; Eagleton, M.J.; et al. Evaluation of Lipoprotein(a) as a Prognostic Marker of Extracoronary Atherosclerotic Vascular Disease Progression. Circulation 2025, 152, 585–598. [Google Scholar] [CrossRef]
  200. Marcovina, S.M.; Koschinsky, M.L. Lipoprotein(a) as a risk factor for coronary artery disease. Am. J. Cardiol. 1998, 82, 57U–66U, discussion 86U. [Google Scholar] [CrossRef] [PubMed]
  201. Reyes-Soffer, G.; Ginsberg, H.N.; Berglund, L.; Duell, P.B.; Heffron, S.P.; Kamstrup, P.R.; Lloyd-Jones, D.M.; Marcovina, S.M.; Yeang, C.; Koschinsky, M.L.; et al. Lipoprotein(a): A Genetically Determined, Causal, and Prevalent Risk Factor for Atherosclerotic Cardiovascular Disease: A Scientific Statement From the American Heart Association. Arter. Thromb. Vasc. Biol. 2022, 42, e48–e60. [Google Scholar] [CrossRef]
  202. Bjornson, E.; Adiels, M.; Taskinen, M.R.; Burgess, S.; Chapman, M.J.; Packard, C.J.; Boren, J. Lipoprotein(a) Is Markedly More Atherogenic Than LDL: An Apolipoprotein B-Based Genetic Analysis. J. Am. Coll. Cardiol. 2024, 83, 385–395. [Google Scholar] [CrossRef]
  203. Tsimikas, S.; Bittner, V. Particle Number and Characteristics of Lipoprotein(a), LDL, and apoB: Perspectives on Contributions to ASCVD. J. Am. Coll. Cardiol. 2024, 83, 396–400. [Google Scholar] [CrossRef] [PubMed]
  204. Bergmark, C.; Dewan, A.; Orsoni, A.; Merki, E.; Miller, E.R.; Shin, M.J.; Binder, C.J.; Horkko, S.; Krauss, R.M.; Chapman, M.J.; et al. A novel function of lipoprotein [a] as a preferential carrier of oxidized phospholipids in human plasma. J. Lipid Res. 2008, 49, 2230–2239. [Google Scholar] [CrossRef]
  205. Tsimikas, S.; Brilakis, E.S.; Miller, E.R.; McConnell, J.P.; Lennon, R.J.; Kornman, K.S.; Witztum, J.L.; Berger, P.B. Oxidized phospholipids, Lp(a) lipoprotein, and coronary artery disease. N. Engl. J. Med. 2005, 353, 46–57. [Google Scholar] [CrossRef] [PubMed]
  206. Sotiriou, S.N.; Orlova, V.V.; Al-Fakhri, N.; Ihanus, E.; Economopoulou, M.; Isermann, B.; Bdeir, K.; Nawroth, P.P.; Preissner, K.T.; Gahmberg, C.G.; et al. Lipoprotein(a) in atherosclerotic plaques recruits inflammatory cells through interaction with Mac-1 integrin. FASEB J. 2006, 20, 559–561. [Google Scholar] [CrossRef]
  207. Schnitzler, J.G.; Hoogeveen, R.M.; Ali, L.; Prange, K.H.M.; Waissi, F.; van Weeghel, M.; Bachmann, J.C.; Versloot, M.; Borrelli, M.J.; Yeang, C.; et al. Atherogenic Lipoprotein(a) Increases Vascular Glycolysis, Thereby Facilitating Inflammation and Leukocyte Extravasation. Circ. Res. 2020, 126, 1346–1359. [Google Scholar] [CrossRef]
  208. Keesler, G.A.; Li, Y.; Skiba, P.J.; Fless, G.M.; Tabas, I. Macrophage foam cell lipoprotein(a)/apoprotein(a) receptor. Cell-surface localization, dependence of induction on new protein synthesis, and ligand specificity. Arter. Thromb. 1994, 14, 1337–1345. [Google Scholar] [CrossRef] [PubMed]
  209. Chaurasia, B.; Summers, S.A. Ceramides in Metabolism: Key Lipotoxic Players. Annu. Rev. Physiol. 2021, 83, 303–330. [Google Scholar] [CrossRef]
  210. Zietzer, A.; Dusing, P.; Reese, L.; Nickenig, G.; Jansen, F. Ceramide Metabolism in Cardiovascular Disease: A Network With High Therapeutic Potential. Arter. Thromb. Vasc. Biol. 2022, 42, 1220–1228. [Google Scholar] [CrossRef] [PubMed]
  211. Summers, S.A. Could Ceramides Become the New Cholesterol? Cell Metab. 2018, 27, 276–280. [Google Scholar] [CrossRef]
  212. Havulinna, A.S.; Sysi-Aho, M.; Hilvo, M.; Kauhanen, D.; Hurme, R.; Ekroos, K.; Salomaa, V.; Laaksonen, R. Circulating Ceramides Predict Cardiovascular Outcomes in the Population-Based FINRISK 2002 Cohort. Arter. Thromb. Vasc. Biol. 2016, 36, 2424–2430. [Google Scholar] [CrossRef]
  213. Meikle, P.J.; Wong, G.; Tsorotes, D.; Barlow, C.K.; Weir, J.M.; Christopher, M.J.; MacIntosh, G.L.; Goudey, B.; Stern, L.; Kowalczyk, A.; et al. Plasma lipidomic analysis of stable and unstable coronary artery disease. Arter. Thromb. Vasc. Biol. 2011, 31, 2723–2732. [Google Scholar] [CrossRef]
  214. Laaksonen, R.; Ekroos, K.; Sysi-Aho, M.; Hilvo, M.; Vihervaara, T.; Kauhanen, D.; Suoniemi, M.; Hurme, R.; Marz, W.; Scharnagl, H.; et al. Plasma ceramides predict cardiovascular death in patients with stable coronary artery disease and acute coronary syndromes beyond LDL-cholesterol. Eur. Heart J. 2016, 37, 1967–1976. [Google Scholar] [CrossRef]
  215. Anroedh, S.; Hilvo, M.; Akkerhuis, K.M.; Kauhanen, D.; Koistinen, K.; Oemrawsingh, R.; Serruys, P.; van Geuns, R.J.; Boersma, E.; Laaksonen, R.; et al. Plasma concentrations of molecular lipid species predict long-term clinical outcome in coronary artery disease patients. J. Lipid Res. 2018, 59, 1729–1737. [Google Scholar] [CrossRef]
  216. Hilvo, M.; Meikle, P.J.; Pedersen, E.R.; Tell, G.S.; Dhar, I.; Brenner, H.; Schottker, B.; Laaperi, M.; Kauhanen, D.; Koistinen, K.M.; et al. Development and validation of a ceramide- and phospholipid-based cardiovascular risk estimation score for coronary artery disease patients. Eur. Heart J. 2020, 41, 371–380. [Google Scholar] [CrossRef] [PubMed]
  217. Gencer, B.; Morrow, D.A.; Braunwald, E.; Goodrich, E.L.; Hilvo, M.; Kauhanen, D.; Sabatine, M.S.; Laaksonen, R.; O’Donoghue, M.L. Plasma ceramide and phospholipid-based risk score and the risk of cardiovascular death in patients after acute coronary syndrome. Eur. J. Prev. Cardiol. 2022, 29, 895–902. [Google Scholar] [CrossRef] [PubMed]
  218. Schissel, S.L.; Tweedie-Hardman, J.; Rapp, J.H.; Graham, G.; Williams, K.J.; Tabas, I. Rabbit aorta and human atherosclerotic lesions hydrolyze the sphingomyelin of retained low-density lipoprotein. Proposed role for arterial-wall sphingomyelinase in subendothelial retention and aggregation of atherogenic lipoproteins. J. Clin. Investig. 1996, 98, 1455–1464. [Google Scholar] [CrossRef]
  219. Cheng, J.M.; Suoniemi, M.; Kardys, I.; Vihervaara, T.; de Boer, S.P.; Akkerhuis, K.M.; Sysi-Aho, M.; Ekroos, K.; Garcia-Garcia, H.M.; Oemrawsingh, R.M.; et al. Plasma concentrations of molecular lipid species in relation to coronary plaque characteristics and cardiovascular outcome: Results of the ATHEROREMO-IVUS study. Atherosclerosis 2015, 243, 560–566. [Google Scholar] [CrossRef]
  220. Li, W.; Yang, X.; Xing, S.; Bian, F.; Yao, W.; Bai, X.; Zheng, T.; Wu, G.; Jin, S. Endogenous ceramide contributes to the transcytosis of oxLDL across endothelial cells and promotes its subendothelial retention in vascular wall. Oxid. Med. Cell Longev. 2014, 2014, 823071. [Google Scholar] [CrossRef] [PubMed]
  221. Walters, M.J.; Wrenn, S.P. Effect of sphingomyelinase-mediated generation of ceramide on aggregation of low-density lipoprotein. Langmuir 2008, 24, 9642–9647. [Google Scholar] [CrossRef]
  222. Devlin, C.M.; Leventhal, A.R.; Kuriakose, G.; Schuchman, E.H.; Williams, K.J.; Tabas, I. Acid sphingomyelinase promotes lipoprotein retention within early atheromata and accelerates lesion progression. Arter. Thromb. Vasc. Biol. 2008, 28, 1723–1730. [Google Scholar] [CrossRef]
  223. Glaros, E.N.; Kim, W.S.; Quinn, C.M.; Jessup, W.; Rye, K.A.; Garner, B. Myriocin slows the progression of established atherosclerotic lesions in apolipoprotein E gene knockout mice. J. Lipid Res. 2008, 49, 324–331. [Google Scholar] [CrossRef]
  224. Gao, D.; Pararasa, C.; Dunston, C.R.; Bailey, C.J.; Griffiths, H.R. Palmitate promotes monocyte atherogenicity via de novo ceramide synthesis. Free Radic. Biol. Med. 2012, 53, 796–806. [Google Scholar] [CrossRef]
  225. Tsimikas, S.; Witztum, J.L. Oxidized phospholipids in cardiovascular disease. Nat. Rev. Cardiol. 2024, 21, 170–191. [Google Scholar] [CrossRef] [PubMed]
  226. Tsimikas, S.; Bergmark, C.; Beyer, R.W.; Patel, R.; Pattison, J.; Miller, E.; Juliano, J.; Witztum, J.L. Temporal increases in plasma markers of oxidized low-density lipoprotein strongly reflect the presence of acute coronary syndromes. J. Am. Coll. Cardiol. 2003, 41, 360–370. [Google Scholar] [CrossRef]
  227. Lee, S.; Birukov, K.G.; Romanoski, C.E.; Springstead, J.R.; Lusis, A.J.; Berliner, J.A. Role of phospholipid oxidation products in atherosclerosis. Circ. Res. 2012, 111, 778–799. [Google Scholar] [CrossRef] [PubMed]
  228. Que, X.; Hung, M.Y.; Yeang, C.; Gonen, A.; Prohaska, T.A.; Sun, X.; Diehl, C.; Maatta, A.; Gaddis, D.E.; Bowden, K.; et al. Oxidized phospholipids are proinflammatory and proatherogenic in hypercholesterolaemic mice. Nature 2018, 558, 301–306. [Google Scholar] [CrossRef]
  229. Boullier, A.; Gillotte, K.L.; Horkko, S.; Green, S.R.; Friedman, P.; Dennis, E.A.; Witztum, J.L.; Steinberg, D.; Quehenberger, O. The binding of oxidized low density lipoprotein to mouse CD36 is mediated in part by oxidized phospholipids that are associated with both the lipid and protein moieties of the lipoprotein. J. Biol. Chem. 2000, 275, 9163–9169. [Google Scholar] [CrossRef]
  230. Gillotte-Taylor, K.; Boullier, A.; Witztum, J.L.; Steinberg, D.; Quehenberger, O. Scavenger receptor class B type I as a receptor for oxidized low density lipoprotein. J. Lipid Res. 2001, 42, 1474–1482. [Google Scholar] [CrossRef]
  231. Kadl, A.; Meher, A.K.; Sharma, P.R.; Lee, M.Y.; Doran, A.C.; Johnstone, S.R.; Elliott, M.R.; Gruber, F.; Han, J.; Chen, W.; et al. Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circ. Res. 2010, 107, 737–746. [Google Scholar] [CrossRef]
  232. Miller, Y.I.; Shyy, J.Y. Context-Dependent Role of Oxidized Lipids and Lipoproteins in Inflammation. Trends Endocrinol. Metab. 2017, 28, 143–152. [Google Scholar] [CrossRef]
  233. Ravandi, A.; Leibundgut, G.; Hung, M.Y.; Patel, M.; Hutchins, P.M.; Murphy, R.C.; Prasad, A.; Mahmud, E.; Miller, Y.I.; Dennis, E.A.; et al. Release and capture of bioactive oxidized phospholipids and oxidized cholesteryl esters during percutaneous coronary and peripheral arterial interventions in humans. J. Am. Coll. Cardiol. 2014, 63, 1961–1971. [Google Scholar] [CrossRef] [PubMed]
  234. Yan, F.X.; Li, H.M.; Li, S.X.; He, S.H.; Dai, W.P.; Li, Y.; Wang, T.T.; Shi, M.M.; Yuan, H.X.; Xu, Z.; et al. The oxidized phospholipid POVPC impairs endothelial function and vasodilation via uncoupling endothelial nitric oxide synthase. J. Mol. Cell Cardiol. 2017, 112, 40–48. [Google Scholar] [CrossRef]
  235. Lu, L.; Ye, Y.; Chen, Y.; Feng, L.; Huang, J.; Liang, Q.; Lan, Z.; Dong, Q.; Liu, X.; Li, Y.; et al. Oxidized phospholipid POVPC contributes to vascular calcification by triggering ferroptosis of vascular smooth muscle cells. FASEB J. 2024, 38, e23592. [Google Scholar] [CrossRef]
  236. Appleton, B.D.; Palmer, S.A.; Smith, H.P.; Stephens, L.E.; Major, A.S. Oxidized Phospholipid oxPAPC Alters Regulatory T-Cell Differentiation and Decreases Their Protective Function in Atherosclerosis in Mice. Arter. Thromb. Vasc. Biol. 2023, 43, 2119–2132. [Google Scholar] [CrossRef] [PubMed]
  237. Di Gioia, M.; Spreafico, R.; Springstead, J.R.; Mendelson, M.M.; Joehanes, R.; Levy, D.; Zanoni, I. Endogenous oxidized phospholipids reprogram cellular metabolism and boost hyperinflammation. Nat. Immunol. 2020, 21, 42–53. [Google Scholar] [CrossRef] [PubMed]
  238. van der Valk, F.M.; Bekkering, S.; Kroon, J.; Yeang, C.; Van den Bossche, J.; van Buul, J.D.; Ravandi, A.; Nederveen, A.J.; Verberne, H.J.; Scipione, C.; et al. Oxidized Phospholipids on Lipoprotein(a) Elicit Arterial Wall Inflammation and an Inflammatory Monocyte Response in Humans. Circulation 2016, 134, 611–624. [Google Scholar] [CrossRef]
  239. De Meyer, G.R.Y.; Zurek, M.; Puylaert, P.; Martinet, W. Programmed death of macrophages in atherosclerosis: Mechanisms and therapeutic targets. Nat. Rev. Cardiol. 2024, 21, 312–325. [Google Scholar] [CrossRef] [PubMed]
  240. Karunakaran, D.; Geoffrion, M.; Wei, L.; Gan, W.; Richards, L.; Shangari, P.; DeKemp, E.M.; Beanlands, R.A.; Perisic, L.; Maegdefessel, L.; et al. Targeting macrophage necroptosis for therapeutic and diagnostic interventions in atherosclerosis. Sci. Adv. 2016, 2, e1600224. [Google Scholar] [CrossRef]
  241. Karunakaran, D.; Nguyen, M.A.; Geoffrion, M.; Vreeken, D.; Lister, Z.; Cheng, H.S.; Otte, N.; Essebier, P.; Wyatt, H.; Kandiah, J.W.; et al. RIPK1 Expression Associates With Inflammation in Early Atherosclerosis in Humans and Can Be Therapeutically Silenced to Reduce NF-kappaB Activation and Atherogenesis in Mice. Circulation 2021, 143, 163–177. [Google Scholar] [CrossRef] [PubMed]
  242. Lin, J.; Li, H.; Yang, M.; Ren, J.; Huang, Z.; Han, F.; Huang, J.; Ma, J.; Zhang, D.; Zhang, Z.; et al. A role of RIP3-mediated macrophage necrosis in atherosclerosis development. Cell Rep. 2013, 3, 200–210. [Google Scholar] [CrossRef] [PubMed]
  243. Rasheed, A.; Robichaud, S.; Nguyen, M.A.; Geoffrion, M.; Wyatt, H.; Cottee, M.L.; Dennison, T.; Pietrangelo, A.; Lee, R.; Lagace, T.A.; et al. Loss of MLKL (Mixed Lineage Kinase Domain-Like Protein) Decreases Necrotic Core but Increases Macrophage Lipid Accumulation in Atherosclerosis. Arter. Thromb. Vasc. Biol. 2020, 40, 1155–1167. [Google Scholar] [CrossRef]
  244. Jiang, M.; Sun, X.; Liu, S.; Tang, Y.; Shi, Y.; Bai, Y.; Wang, Y.; Yang, Q.; Yang, Q.; Jiang, W.; et al. Caspase-11-Gasdermin D-Mediated Pyroptosis Is Involved in the Pathogenesis of Atherosclerosis. Front. Pharmacol. 2021, 12, 657486. [Google Scholar] [CrossRef]
  245. Shi, X.; Xie, W.L.; Kong, W.W.; Chen, D.; Qu, P. Expression of the NLRP3 Inflammasome in Carotid Atherosclerosis. J. Stroke Cerebrovasc. Dis. 2015, 24, 2455–2466. [Google Scholar] [CrossRef]
  246. Rajamaki, K.; Mayranpaa, M.I.; Risco, A.; Tuimala, J.; Nurmi, K.; Cuenda, A.; Eklund, K.K.; Oorni, K.; Kovanen, P.T. p38delta MAPK: A Novel Regulator of NLRP3 Inflammasome Activation With Increased Expression in Coronary Atherogenesis. Arter. Thromb. Vasc. Biol. 2016, 36, 1937–1946. [Google Scholar] [CrossRef]
  247. Zheng, F.; Xing, S.; Gong, Z.; Xing, Q. NLRP3 inflammasomes show high expression in aorta of patients with atherosclerosis. Heart Lung Circ. 2013, 22, 746–750. [Google Scholar] [CrossRef]
  248. Duewell, P.; Kono, H.; Rayner, K.J.; Sirois, C.M.; Vladimer, G.; Bauernfeind, F.G.; Abela, G.S.; Franchi, L.; Nunez, G.; Schnurr, M.; et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010, 464, 1357–1361. [Google Scholar] [CrossRef] [PubMed]
  249. Balasubramanian, A.; Hsu, A.Y.; Ghimire, L.; Tahir, M.; Devant, P.; Fontana, P.; Du, G.; Liu, X.; Fabin, D.; Kambara, H.; et al. The palmitoylation of gasdermin D directs its membrane translocation and pore formation during pyroptosis. Sci. Immunol. 2024, 9, eadn1452. [Google Scholar] [CrossRef]
  250. Du, G.; Healy, L.B.; David, L.; Walker, C.; El-Baba, T.J.; Lutomski, C.A.; Goh, B.; Gu, B.; Pi, X.; Devant, P.; et al. ROS-dependent S-palmitoylation activates cleaved and intact gasdermin D. Nature 2024, 630, 437–446. [Google Scholar] [CrossRef]
  251. Yalcinkaya, M.; Fotakis, P.; Liu, W.; Endo-Umeda, K.; Dou, H.; Abramowicz, S.; Xiao, T.; Libby, P.; Wang, N.; Tall, A.R.; et al. Cholesterol accumulation in macrophages drives NETosis in atherosclerotic plaques via IL-1beta secretion. Cardiovasc. Res. 2023, 119, 969–981. [Google Scholar] [CrossRef]
  252. Fuhrman, B.; Oiknine, J.; Aviram, M. Iron induces lipid peroxidation in cultured macrophages, increases their ability to oxidatively modify LDL, and affects their secretory properties. Atherosclerosis 1994, 111, 65–78. [Google Scholar] [CrossRef]
  253. Yuan, X.M.; Anders, W.L.; Olsson, A.G.; Brunk, U.T. Iron in human atheroma and LDL oxidation by macrophages following erythrophagocytosis. Atherosclerosis 1996, 124, 61–73. [Google Scholar] [CrossRef]
  254. Wang, L.J.; Lee, T.S.; Lee, F.Y.; Pai, R.C.; Chau, L.Y. Expression of heme oxygenase-1 in atherosclerotic lesions. Am. J. Pathol. 1998, 152, 711–720. [Google Scholar]
  255. Yuan, X.M.; Li, W.; Baird, S.K.; Carlsson, M.; Melefors, O. Secretion of ferritin by iron-laden macrophages and influence of lipoproteins. Free Radic. Res. 2004, 38, 1133–1142. [Google Scholar] [CrossRef]
  256. Bai, T.; Li, M.; Liu, Y.; Qiao, Z.; Wang, Z. Inhibition of ferroptosis alleviates atherosclerosis through attenuating lipid peroxidation and endothelial dysfunction in mouse aortic endothelial cell. Free Radic. Biol. Med. 2020, 160, 92–102. [Google Scholar] [CrossRef] [PubMed]
  257. Ward, N.C.; Watts, G.F.; Eckel, R.H. Statin Toxicity. Circ. Res. 2019, 124, 328–350. [Google Scholar] [CrossRef] [PubMed]
  258. Chou, R.; Cantor, A.; Dana, T.; Wagner, J.; Ahmed, A.Y.; Fu, R.; Ferencik, M. Statin Use for the Primary Prevention of Cardiovascular Disease in Adults: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA 2022, 328, 754–771. [Google Scholar] [CrossRef] [PubMed]
  259. Force, U.S.P.S.T.; Mangione, C.M.; Barry, M.J.; Nicholson, W.K.; Cabana, M.; Chelmow, D.; Coker, T.R.; Davis, E.M.; Donahue, K.E.; Jaen, C.R.; et al. Statin Use for the Primary Prevention of Cardiovascular Disease in Adults: US Preventive Services Task Force Recommendation Statement. JAMA 2022, 328, 746–753. [Google Scholar] [CrossRef]
  260. Razavi, A.C.; Mehta, A.; Sperling, L.S. Statin therapy for the primary prevention of cardiovascular disease: Pros. Atherosclerosis 2022, 356, 41–45. [Google Scholar] [CrossRef]
  261. Fitchett, D.H.; Hegele, R.A.; Verma, S. Cardiology patient page. Statin intolerance. Circ. 2015, 131, e389–e391. [Google Scholar] [CrossRef]
  262. Bytyci, I.; Penson, P.E.; Mikhailidis, D.P.; Wong, N.D.; Hernandez, A.V.; Sahebkar, A.; Thompson, P.D.; Mazidi, M.; Rysz, J.; Pella, D.; et al. Prevalence of statin intolerance: A meta-analysis. Eur. Heart J. 2022, 43, 3213–3223. [Google Scholar] [CrossRef]
  263. Keaney, J.F., Jr. Bempedoic Acid and the Prevention of Cardiovascular Disease. N. Engl. J. Med. 2023, 388, 1427–1430. [Google Scholar] [CrossRef]
  264. Ray, K.K.; Bays, H.E.; Catapano, A.L.; Lalwani, N.D.; Bloedon, L.T.; Sterling, L.R.; Robinson, P.L.; Ballantyne, C.M.; Trial, C.H. Safety and Efficacy of Bempedoic Acid to Reduce LDL Cholesterol. N. Engl. J. Med. 2019, 380, 1022–1032. [Google Scholar] [CrossRef]
  265. Nissen, S.E.; Lincoff, A.M.; Brennan, D.; Ray, K.K.; Mason, D.; Kastelein, J.J.P.; Thompson, P.D.; Libby, P.; Cho, L.; Plutzky, J.; et al. Bempedoic Acid and Cardiovascular Outcomes in Statin-Intolerant Patients. N. Engl. J. Med. 2023, 388, 1353–1364. [Google Scholar] [CrossRef]
  266. Visser, M.E.; Wagener, G.; Baker, B.F.; Geary, R.S.; Donovan, J.M.; Beuers, U.H.; Nederveen, A.J.; Verheij, J.; Trip, M.D.; Basart, D.C.; et al. Mipomersen, an apolipoprotein B synthesis inhibitor, lowers low-density lipoprotein cholesterol in high-risk statin-intolerant patients: A randomized, double-blind, placebo-controlled trial. Eur. Heart J. 2012, 33, 1142–1149. [Google Scholar] [CrossRef] [PubMed]
  267. Stein, E.A.; Dufour, R.; Gagne, C.; Gaudet, D.; East, C.; Donovan, J.M.; Chin, W.; Tribble, D.L.; McGowan, M. Apolipoprotein B synthesis inhibition with mipomersen in heterozygous familial hypercholesterolemia: Results of a randomized, double-blind, placebo-controlled trial to assess efficacy and safety as add-on therapy in patients with coronary artery disease. Circulation 2012, 126, 2283–2292. [Google Scholar] [CrossRef]
  268. Santos, R.D.; Raal, F.J.; Catapano, A.L.; Witztum, J.L.; Steinhagen-Thiessen, E.; Tsimikas, S. Mipomersen, an antisense oligonucleotide to apolipoprotein B-100, reduces lipoprotein(a) in various populations with hypercholesterolemia: Results of 4 phase III trials. Arter. Thromb. Vasc. Biol. 2015, 35, 689–699. [Google Scholar] [CrossRef]
  269. Chambergo-Michilot, D.; Alur, A.; Kulkarni, S.; Agarwala, A. Mipomersen in Familial Hypercholesterolemia: An Update on Health-Related Quality of Life and Patient-Reported Outcomes. Vasc. Health Risk Manag. 2022, 18, 73–80. [Google Scholar] [CrossRef]
  270. Seidah, N.G.; Prat, A. The Multifaceted Biology of PCSK9. Endocr. Rev. 2022, 43, 558–582. [Google Scholar] [CrossRef]
  271. Nie, W.; Yue, Y.; Hu, J. PCSK9 inhibitors in the management of atherosclerotic cardiovascular disease: Current clinical trials and future directions. Atherosclerosis 2024, 399, 119043. [Google Scholar] [CrossRef]
  272. Sabatine, M.S.; Giugliano, R.P.; Keech, A.C.; Honarpour, N.; Wiviott, S.D.; Murphy, S.A.; Kuder, J.F.; Wang, H.; Liu, T.; Wasserman, S.M.; et al. Evolocumab and Clinical Outcomes in Patients with Cardiovascular Disease. N. Engl. J. Med. 2017, 376, 1713–1722. [Google Scholar] [CrossRef] [PubMed]
  273. Schwartz, G.G.; Steg, P.G.; Szarek, M.; Bhatt, D.L.; Bittner, V.A.; Diaz, R.; Edelberg, J.M.; Goodman, S.G.; Hanotin, C.; Harrington, R.A.; et al. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N. Engl. J. Med. 2018, 379, 2097–2107. [Google Scholar] [CrossRef] [PubMed]
  274. Ray, K.K.; Wright, R.S.; Kallend, D.; Koenig, W.; Leiter, L.A.; Raal, F.J.; Bisch, J.A.; Richardson, T.; Jaros, M.; Wijngaard, P.L.J.; et al. Two Phase 3 Trials of Inclisiran in Patients with Elevated LDL Cholesterol. N. Engl. J. Med. 2020, 382, 1507–1519. [Google Scholar] [CrossRef] [PubMed]
  275. Natarajan, P.; Kathiresan, S. PCSK9 Inhibitors. Cell 2016, 165, 1037. [Google Scholar] [CrossRef]
  276. Raal, F.J.; Kallend, D.; Ray, K.K.; Turner, T.; Koenig, W.; Wright, R.S.; Wijngaard, P.L.J.; Curcio, D.; Jaros, M.J.; Leiter, L.A.; et al. Inclisiran for the Treatment of Heterozygous Familial Hypercholesterolemia. N. Engl. J. Med. 2020, 382, 1520–1530. [Google Scholar] [CrossRef]
  277. Mazer, C.D.; Moody, A.R.; Teoh, H.; Quan, A.; Elituv, R.; Vivekanandan, T.; Li, P.; Thorpe, K.E.; Lafreniere-Roula, M.; Verma, R.; et al. SLICE-CEA CardioLink-8: A Randomized Trial of Evolocumab on Carotid Artery Atherosclerotic Plaque Characteristics in Asymptomatic High-Risk Carotid Stenosis. Circulation 2025, 151, 351–355. [Google Scholar] [CrossRef]
  278. Bittner, V.A.; Szarek, M.; Aylward, P.E.; Bhatt, D.L.; Diaz, R.; Edelberg, J.M.; Fras, Z.; Goodman, S.G.; Halvorsen, S.; Hanotin, C.; et al. Effect of Alirocumab on Lipoprotein(a) and Cardiovascular Risk After Acute Coronary Syndrome. J. Am. Coll. Cardiol. 2020, 75, 133–144. [Google Scholar] [CrossRef]
  279. Tsimikas, S. Lipoprotein(a) in the Year 2024: A Look Back and a Look Ahead. Arter. Thromb. Vasc. Biol. 2024, 44, 1485–1490. [Google Scholar] [CrossRef] [PubMed]
  280. Witztum, J.L.; Gaudet, D.; Freedman, S.D.; Alexander, V.J.; Digenio, A.; Williams, K.R.; Yang, Q.; Hughes, S.G.; Geary, R.S.; Arca, M.; et al. Volanesorsen and Triglyceride Levels in Familial Chylomicronemia Syndrome. N. Engl. J. Med. 2019, 381, 531–542. [Google Scholar] [CrossRef] [PubMed]
  281. Bergmark, B.A.; Marston, N.A.; Prohaska, T.A.; Alexander, V.J.; Zimerman, A.; Moura, F.A.; Murphy, S.A.; Goodrich, E.L.; Zhang, S.; Gaudet, D.; et al. Olezarsen for Hypertriglyceridemia in Patients at High Cardiovascular Risk. N. Engl. J. Med. 2024, 390, 1770–1780. [Google Scholar] [CrossRef] [PubMed]
  282. Bergmark, B.A.; Marston, N.A.; Prohaska, T.A.; Alexander, V.J.; Zimerman, A.; Moura, F.A.; Kang, Y.M.; Weinland, J.; Murphy, S.A.; Goodrich, E.L.; et al. Targeting APOC3 with Olezarsen in Moderate Hypertriglyceridemia. N. Engl. J. Med. 2025. [Google Scholar] [CrossRef]
  283. Nordestgaard, A.T.; Tybjaerg-Hansen, A.; Mansbach, H.; Kersten, S.; Nordestgaard, B.G.; Rosenson, R.S. Target Populations for Novel Triglyceride-Lowering Therapies. J. Am. Coll. Cardiol. 2025, 85, 1876–1897. [Google Scholar] [CrossRef]
  284. Raal, F.J.; Rosenson, R.S.; Reeskamp, L.F.; Hovingh, G.K.; Kastelein, J.J.P.; Rubba, P.; Ali, S.; Banerjee, P.; Chan, K.C.; Gipe, D.A.; et al. Evinacumab for Homozygous Familial Hypercholesterolemia. N. Engl. J. Med. 2020, 383, 711–720. [Google Scholar] [CrossRef]
  285. Rosenson, R.S.; Burgess, L.J.; Ebenbichler, C.F.; Baum, S.J.; Stroes, E.S.G.; Ali, S.; Khilla, N.; Hamlin, R.; Pordy, R.; Dong, Y.; et al. Evinacumab in Patients with Refractory Hypercholesterolemia. N. Engl. J. Med. 2020, 383, 2307–2319. [Google Scholar] [CrossRef]
  286. Rohatgi, A.; Westerterp, M.; von Eckardstein, A.; Remaley, A.; Rye, K.A. HDL in the 21st Century: A Multifunctional Roadmap for Future HDL Research. Circulation 2021, 143, 2293–2309. [Google Scholar] [CrossRef]
  287. Tall, A.R.; Rader, D.J. Trials and Tribulations of CETP Inhibitors. Circ. Res. 2018, 122, 106–112. [Google Scholar] [CrossRef]
  288. Armitage, J.; Holmes, M.V.; Preiss, D. Cholesteryl Ester Transfer Protein Inhibition for Preventing Cardiovascular Events: JACC Review Topic of the Week. J. Am. Coll. Cardiol. 2019, 73, 477–487. [Google Scholar] [CrossRef] [PubMed]
  289. Group, H.T.R.C.; Bowman, L.; Hopewell, J.C.; Chen, F.; Wallendszus, K.; Stevens, W.; Collins, R.; Wiviott, S.D.; Cannon, C.P.; Braunwald, E.; et al. Effects of Anacetrapib in Patients with Atherosclerotic Vascular Disease. N. Engl. J. Med. 2017, 377, 1217–1227. [Google Scholar] [CrossRef]
  290. Krause, B.R.; Remaley, A.T. Reconstituted HDL for the acute treatment of acute coronary syndrome. Curr. Opin. Lipidol. 2013, 24, 480–486. [Google Scholar] [CrossRef] [PubMed]
  291. Gibson, C.M.; Duffy, D.; Korjian, S.; Bahit, M.C.; Chi, G.; Alexander, J.H.; Lincoff, A.M.; Heise, M.; Tricoci, P.; Deckelbaum, L.I.; et al. Apolipoprotein A1 Infusions and Cardiovascular Outcomes after Acute Myocardial Infarction. N. Engl. J. Med. 2024, 390, 1560–1571. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Modified cholesterol-rich lipoproteins drive vascular and immune-mediated dysfunctions that potentiate atherogenesis. Under homeostatic conditions, circulating monocytes roll along an intact endothelial barrier. However, in response to the hyperlipidemic environment that characterizes atherosclerosis, endothelial defects facilitate monocyte extravasation and lipoprotein accumulation in the sub-endothelial space of the arterial wall. Here, monocytes encounter vascular-produced macrophage colony-stimulating factor (M-CSF) to differentiate into the macrophages that engulf the modified lipoproteins via scavenger receptors. These lipoproteins are trafficked to the lysosome and metabolized by lysosomal acid lipase (LAL) to liberate free cholesterol. Free cholesterol can then be esterified to fatty acids in the endoplasmic reticulum by acyl-coenzyme A:cholesterol acyltransferases 1 (ACAT1) to form lipid droplets. Alternatively, free cholesterol can be metabolized to generate oxysterols, which in turn activate the liver x receptors (LXRs) to induce transcription of the cholesterol transporters ATP-binding cassette A1 and G1 (ABCA1/G1). Lipid droplets can be broken down by autophagy to produce free cholesterol, which is removed from macrophages to apolipoprotein A1 (Apo-A1) and high-density lipoprotein (HDL) by ABCA1 and ABCG1, respectively. In atherogenic foam cells, accumulation of free cholesterol leads to the generation of cholesterol crystals or oxysterols that induce inflammation. These cells are also characterized by endoplasmic reticulum stress and apoptosis. During the late stages of atherosclerosis, macrophage foam cells undergo non-apoptotic cell death, leading to the necrotic core, including pore formation and cell lysis mediated by ferroptosis, pyroptosis, and necroptosis.
Figure 1. Modified cholesterol-rich lipoproteins drive vascular and immune-mediated dysfunctions that potentiate atherogenesis. Under homeostatic conditions, circulating monocytes roll along an intact endothelial barrier. However, in response to the hyperlipidemic environment that characterizes atherosclerosis, endothelial defects facilitate monocyte extravasation and lipoprotein accumulation in the sub-endothelial space of the arterial wall. Here, monocytes encounter vascular-produced macrophage colony-stimulating factor (M-CSF) to differentiate into the macrophages that engulf the modified lipoproteins via scavenger receptors. These lipoproteins are trafficked to the lysosome and metabolized by lysosomal acid lipase (LAL) to liberate free cholesterol. Free cholesterol can then be esterified to fatty acids in the endoplasmic reticulum by acyl-coenzyme A:cholesterol acyltransferases 1 (ACAT1) to form lipid droplets. Alternatively, free cholesterol can be metabolized to generate oxysterols, which in turn activate the liver x receptors (LXRs) to induce transcription of the cholesterol transporters ATP-binding cassette A1 and G1 (ABCA1/G1). Lipid droplets can be broken down by autophagy to produce free cholesterol, which is removed from macrophages to apolipoprotein A1 (Apo-A1) and high-density lipoprotein (HDL) by ABCA1 and ABCG1, respectively. In atherogenic foam cells, accumulation of free cholesterol leads to the generation of cholesterol crystals or oxysterols that induce inflammation. These cells are also characterized by endoplasmic reticulum stress and apoptosis. During the late stages of atherosclerosis, macrophage foam cells undergo non-apoptotic cell death, leading to the necrotic core, including pore formation and cell lysis mediated by ferroptosis, pyroptosis, and necroptosis.
Lipidology 02 00017 g001
Figure 2. Lipid-based approaches to treat ASCVD patients. Low-density lipoprotein (LDL) cholesterol is taken up from the circulation by apolipoprotein-B100 (Apo-B100) binding to the LDL receptor (LDLR). Upon binding and entry into hepatocytes via the endosomal system, LDL is transported to the lysosome for metabolism, and LDLR is recycled to the plasma membrane to promote further LDL uptake. However, when proprotein convertase subtilisin/kexin type 9 (PCSK9) is bound to LDLR, it prevents LDLR transport back to the plasma membrane and promotes its lysosomal degradation, thereby blunting LDL uptake. In addition to uptake from the circulation, hepatic cholesterol content is influenced by cholesterol biosynthesis, mediated by a multistep conversion process beginning with acetyl-CoA. This involves many enzymes that are the subject of clinical intervention, including ATP citrate lyase (ACLY), which is inhibited by bempedoic acid, and the rate-limiting enzyme of this process, HMG-CoA reductase, which is the target of statins. Lipids within hepatocytes are packaged into lipoproteins, which express apolipoproteins to facilitate their uptake. Very low-density lipoprotein (VLDL) is broken down, and triglycerides are released by lipoprotein lipase in the muscle and adipose tissue. Within the plaque, cholesterol is removed by cholesterol efflux transporters to apolipoprotein A1 (Apo-A1) and high-density lipoprotein (HDL), which is taken up by the liver via Apo-A1 binding to scavenger receptor B1 (SR-B1) expressed on hepatocytes. In the circulation, cholesteryl ester transfer protein (CETP) exchanges cholesterol on HDL for cholesterol on LDL and VLDL. All clinical targets are indicated in red.
Figure 2. Lipid-based approaches to treat ASCVD patients. Low-density lipoprotein (LDL) cholesterol is taken up from the circulation by apolipoprotein-B100 (Apo-B100) binding to the LDL receptor (LDLR). Upon binding and entry into hepatocytes via the endosomal system, LDL is transported to the lysosome for metabolism, and LDLR is recycled to the plasma membrane to promote further LDL uptake. However, when proprotein convertase subtilisin/kexin type 9 (PCSK9) is bound to LDLR, it prevents LDLR transport back to the plasma membrane and promotes its lysosomal degradation, thereby blunting LDL uptake. In addition to uptake from the circulation, hepatic cholesterol content is influenced by cholesterol biosynthesis, mediated by a multistep conversion process beginning with acetyl-CoA. This involves many enzymes that are the subject of clinical intervention, including ATP citrate lyase (ACLY), which is inhibited by bempedoic acid, and the rate-limiting enzyme of this process, HMG-CoA reductase, which is the target of statins. Lipids within hepatocytes are packaged into lipoproteins, which express apolipoproteins to facilitate their uptake. Very low-density lipoprotein (VLDL) is broken down, and triglycerides are released by lipoprotein lipase in the muscle and adipose tissue. Within the plaque, cholesterol is removed by cholesterol efflux transporters to apolipoprotein A1 (Apo-A1) and high-density lipoprotein (HDL), which is taken up by the liver via Apo-A1 binding to scavenger receptor B1 (SR-B1) expressed on hepatocytes. In the circulation, cholesteryl ester transfer protein (CETP) exchanges cholesterol on HDL for cholesterol on LDL and VLDL. All clinical targets are indicated in red.
Lipidology 02 00017 g002
Table 1. Clinical values for circulating lipids in adults 1.
Table 1. Clinical values for circulating lipids in adults 1.
Circulating LipidsNormal/Optimal Ranges for Adults
LDL cholesterol<100 mg/dL (2.6 mM)
HDL cholesterol>40 mg/dL (1.0 mM)
Fasting triglycerides<150 mg/dL (1.7 mM)
Lp(a) *<30 mg/dL (62 nM)
1 Data compiled from [11,32,33,34]. * Need for standardization in Lp(a) measurements contribute to the variability in recommended values [35]. Concentrations calculated according to [36].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Steinbeck, J.E.; Iannotti, R.A.; Rasheed, A. Dysregulated Lipid Metabolism as a Central Driver of Atherosclerotic Plaque Pathology. Lipidology 2025, 2, 17. https://doi.org/10.3390/lipidology2040017

AMA Style

Steinbeck JE, Iannotti RA, Rasheed A. Dysregulated Lipid Metabolism as a Central Driver of Atherosclerotic Plaque Pathology. Lipidology. 2025; 2(4):17. https://doi.org/10.3390/lipidology2040017

Chicago/Turabian Style

Steinbeck, Julia Emily, Rachel Anne Iannotti, and Adil Rasheed. 2025. "Dysregulated Lipid Metabolism as a Central Driver of Atherosclerotic Plaque Pathology" Lipidology 2, no. 4: 17. https://doi.org/10.3390/lipidology2040017

APA Style

Steinbeck, J. E., Iannotti, R. A., & Rasheed, A. (2025). Dysregulated Lipid Metabolism as a Central Driver of Atherosclerotic Plaque Pathology. Lipidology, 2(4), 17. https://doi.org/10.3390/lipidology2040017

Article Metrics

Back to TopTop